US20210221910A1 - Anti-muc1 antibody - Google Patents

Anti-muc1 antibody Download PDF

Info

Publication number
US20210221910A1
US20210221910A1 US17/055,303 US201917055303A US2021221910A1 US 20210221910 A1 US20210221910 A1 US 20210221910A1 US 201917055303 A US201917055303 A US 201917055303A US 2021221910 A1 US2021221910 A1 US 2021221910A1
Authority
US
United States
Prior art keywords
cancer
amino acid
antibody
seq
acid sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/055,303
Inventor
Johanna GELLERT
Anke Flechner
Doreen Weigelt
Antje Danielczyk
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Glycotope GmbH
Original Assignee
Glycotope GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glycotope GmbH filed Critical Glycotope GmbH
Assigned to GLYCOTOPE GMBH reassignment GLYCOTOPE GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DANIELCZYK, ANTJE, FLECHNER, Anke, Gellert, Johanna, WEIGELT, Doreen
Publication of US20210221910A1 publication Critical patent/US20210221910A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • C07K16/3092Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties against tumour-associated mucins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention pertains to the field of antibodies.
  • a mutated anti-MUC1 antibody with increased antigen binding affinity is provided.
  • a mutated version of the humanized antibody PankoMab is provided wherein asparagine 57 of the heavy chain variable region is substituted by another amino acid. Thereby, the glycosylation site in the CDR2 region is deleted and the antigen binding affinity is increased.
  • the present invention is directed to the therapeutic and diagnostic use of this antibody and to methods of producing such antibodies.
  • Antibodies against tumor-associated antigens are widely used therapeutics against cancers.
  • Today, many anti-cancer antibodies are approved for human therapy. Some of these antibodies act by blocking certain signaling pathways which are critical for survival or proliferation of specific cancer cells.
  • Other anti-cancer antibodies activate the patient's immune response against the targeted cancer cells, for example by initiating antibody-dependent cellular cytotoxicity (ADCC) via natural killer cells. This mechanism is induced by binding of the antibody's Fc part to Fc receptors on the immune cells.
  • ADCC antibody-dependent cellular cytotoxicity
  • Mucins are a family of high molecular weight, heavily glycosylated proteins produced by many epithelial tissues in vertebrates. They can be subdivided into mucin proteins which are membrane-bound due to the presence of a hydrophobic membrane-spanning domain that favors retention in the plasma membrane, and mucins which are secreted onto mucosal surfaces or secreted to become a component of saliva.
  • the human mucin protein family consists of many family members, including membrane bound MUC1.
  • Mucins are also overexpressed in lung diseases such as asthma, bronchitis, chronic obstructive pulmonary disease or cystic fibrosis.
  • MUC1 and MUC4 Two membrane mucins, MUC1 and MUC4 have been extensively studied in relation to their pathological implication in the disease process.
  • mucins are also being investigated for their potential as diagnostic markers.
  • Several antibodies directed against mucin proteins (Clin. Cancer Res., 2011 Nov. 1;17(21):6822-30, PLoS One, 2011 Jan. 14;6(1):e15921), in particular MUC1, are known in the art. However, their therapeutic efficacy could still be improved.
  • the present inventors have found that deleting the glycosylation site in the heavy chain variable region of the anti-MUC1 antibody PankoMab did not abolish antigen binding, but rather unexpectedly increased the antigen affinity of the antibody. This was in particular surprising as the glycosylation site is located in the second complementarity-determining region of the heavy chain variable region (CDR-H2).
  • the CDRs are those regions of an antibody which are directly involved in antigen binding and provide the contact to the epitope. Therefore, generally modifying the amino acids of a CDR is expected to be detrimental to the antigen binding affinity.
  • the humanized PankoMab antibody additionally comprises a glycosylation site in CDR-H2, which carries a large carbohydrate structure. This carbohydrate structure is present directly at the binding interface to the antigen and hence, was considered to be involved in antigen binding.
  • the PankoMab variant (N54Q) wherein the glycosylation site is deleted by substituting the amino acid carrying the carbohydrate structure exhibits an increased antigen binding affinity.
  • the present invention is directed to an antibody capable of binding to MUC1, which comprises
  • the present invention provides a nucleic acid encoding the antibody according to the invention. Furthermore, in a third aspect an expression cassette or vector comprising the nucleic acid according to the invention and a promoter operatively connected with said nucleic acid and, in a fourth aspect, a host cell comprising the nucleic acid or the expression cassette or vector according to the invention are provided.
  • the present invention provides a conjugate comprising the antibody according to the invention conjugated to a further agent.
  • the present invention is directed to a composition
  • a composition comprising the antibody according to the invention, the nucleic acid according to the invention, the expression cassette or vector according to the invention, the host cell according to the invention, or the conjugate according to the invention.
  • the invention provides the antibody, the nucleic acid, the expression cassette or vector, the host cell, the composition or the conjugate according to the invention for use in medicine, in particular in the treatment, prevention or diagnosis of cancer.
  • the invention provides a method of increasing the MUC1 binding affinity of an antibody comprising
  • the method comprising the step of substituting the amino acid residue at position 8 of CDR-H2 with any amino acid residue except asparagine, resulting in CDR-H2 having the amino acid sequence of SEQ ID NO: 2.
  • the invention provides a method of producing an antibody with increased MUC1 binding affinity, comprising
  • the present invention provides a method for treating cancer in a subject in need thereof comprising, administering to the subject with cancer a therapeutically effective amount of the antibody according to the invention, the nucleic acid according to the invention, the expression cassette or vector according to the invention, or the host cell according to the invention.
  • kits or devices comprising the antibody according to the invention, and associated methods that are useful in the diagnosis, detecting or monitoring of MUC1 associated disorders such as cancer.
  • antibody in particular refers to a protein comprising at least two heavy chains and two light chains connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (V H ) and a heavy chain constant region (C H ).
  • Each light chain is comprised of a light chain variable region (V L ) and a light chain constant region (C L ).
  • the heavy chain-constant region comprises three or—in the case of antibodies of the IgM- or IgE-type—four heavy chain-constant domains (C H1 , C H2 , C H3 and C H4 ) wherein the first constant domain C H1 is adjacent to the variable region and may be connected to the second constant domain CH 2 by a hinge region.
  • the light chain-constant region consists only of one constant domain.
  • the variable regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR), wherein each variable region comprises three CDRs and four FRs.
  • CDRs complementarity determining regions
  • FR framework regions
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the heavy chain constant regions may be of any type such as ⁇ -, ⁇ -, ⁇ -, ⁇ - or ⁇ -type heavy chains.
  • the heavy chain of the antibody is a ⁇ -chain.
  • the light chain constant region may also be of any type such as ⁇ - or ⁇ -type light chains.
  • the light chain of the antibody is a ⁇ -chain.
  • ⁇ - ( ⁇ -, ⁇ -, ⁇ - or ⁇ -) type heavy chain and “ ⁇ - ( ⁇ -) type light chain” refer to antibody heavy chains or antibody light chains, respectively, which have constant region amino acid sequences derived from naturally occurring heavy or light chain constant region amino acid sequences, especially human heavy or light chain constant region amino acid sequences.
  • the amino acid sequence of the constant domains of a ⁇ -type (especially ⁇ 1-type) heavy chain is at least 95%, especially at least 98%, identical to the amino acid sequence of the constant domains of a human ⁇ (especially the human ⁇ 1) antibody heavy chain.
  • the amino acid sequence of the constant domain of a ⁇ -type light chain is in particular at least 95%, especially at least 98%, identical to the amino acid sequence of the constant domain of the human K antibody light chain.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system.
  • the antibody can be e.g. a humanized, human or chimeric antibody.
  • the antigen-binding portion of an antibody usually refers to full length or one or more fragments of an antibody that retains the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments of an antibody examples include a Fab fragment, a monovalent fragment consisting of the V L , V H , C L and C H1 domains; a F(ab) 2 fragment, a bivalent fragment comprising two Fab fragments, each of which binds to the same antigen, linked by a disulfide bridge at the hinge region; a Fd fragment consisting of the V H and C H1 domains; a Fv fragment consisting of the V L and V H domains of a single arm of an antibody; and a dAb fragment, which consists of a V H domain.
  • the “Fab part” of an antibody in particular refers to a part of the antibody comprising the heavy and light chain variable regions (V H and V L ) and the first domains of the heavy and light chain constant regions (C H1 and C L ). In cases where the antibody does not comprise all of these regions, then the term “Fab part” only refers to those of the regions V H , V L , C H1 and C L which are present in the antibody.
  • “Fab part” refers to that part of an antibody corresponding to the fragment obtained by digesting a natural antibody with papain which contains the antigen binding activity of the antibody.
  • the Fab part of an antibody encompasses the antigen binding site or antigen binding ability thereof.
  • the Fab part comprises at least the V H region of the antibody.
  • the “Fc part” of an antibody in particular refers to a part of the antibody comprising the heavy chain constant regions 2, 3 and—where applicable—4 (C H2 , C H3 and C H4 ).
  • the Fc part comprises two of each of these regions.
  • the term “Fc part” only refers to those of the regions O H2 , H 3 and C H4 which are present in the antibody.
  • the Fc part comprises at least the C H2 region of the antibody.
  • “Fc part” refers to that part of an antibody corresponding to the fragment obtained by digesting a natural antibody with papain which does not contain the antigen binding activity of the antibody.
  • the Fc part of an antibody is capable of binding to the Fc receptor and thus, e.g. comprises an Fc receptor binding site or an Fc receptor binding ability.
  • antibody and antibody construct refer in certain embodiments to a population of antibodies or antibody constructs, respectively, of the same kind. In particular, all antibodies or antibody constructs of the population exhibit the features used for defining the antibody or antibody construct. In certain embodiments, all antibodies or antibody constructs in the population have the same amino acid sequence. Reference to a specific kind of antibody, such as an antibody capable of specifically binding to MUC1, in particular refers to a population of this kind of antibody.
  • antibody as used herein also includes fragments and derivatives of said antibody.
  • a “fragment or derivative” of an antibody in particular is a protein or glycoprotein which is derived from said antibody and is capable of binding to the same antigen, in particular to the same epitope as the antibody.
  • a fragment or derivative of an antibody herein generally refers to a functional fragment or derivative.
  • the fragment or derivative of an antibody comprises a heavy chain variable region. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody or derivatives thereof.
  • fragments of an antibody include (i) Fab fragments, monovalent fragments consisting of the variable region and the first constant domain of each the heavy and the light chain; (ii) F(ab) 2 fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) Fd fragments consisting of the variable region and the first constant domain C H1 of the heavy chain; (iv) Fv fragments consisting of the heavy chain and light chain variable region of a single arm of an antibody; (v) scFv fragments, Fv fragments consisting of a single polypeptide chain; (vi) (Fv) 2 fragments consisting of two Fv fragments covalently linked together; (vii) a heavy chain variable domain; and (viii) multibodies consisting of a heavy chain variable region and a light chain variable region covalently linked together in such a manner that association of the heavy chain and light chain variable regions can only occur intermolecular but not intramolecular.
  • Derivatives of an antibody in particular include antibodies which bind to or compete with the same antigen as the parent antibody, but which have a different amino acid sequence than the parent antibody from which it is derived. These antibody fragments and derivatives are obtained using conventional techniques known to those with skill in the art.
  • a target amino acid sequence is “derived” from or “corresponds” to a reference amino acid sequence if the target amino acid sequence shares a homology or identity over its entire length with a corresponding part of the reference amino acid sequence of at least 75%, more preferably at least 80%, at least 85%, at least 90%, at least 93%, at least 95%, at least 97%, at least 98% or at least 99%.
  • the “corresponding part” means that, for example, framework region 1 of a heavy chain variable region (FRH1) of a target antibody corresponds to framework region 1 of the heavy chain variable region of the reference antibody.
  • a target amino acid sequence which is “derived” from or “corresponds” to a reference amino acid sequence is 100% homologous, or in particular 100% identical, over its entire length with a corresponding part of the reference amino acid sequence.
  • a “homology” or “identity” of an amino acid sequence or nucleotide sequence is preferably determined according to the invention over the entire length of the reference sequence or over the entire length of the corresponding part of the reference sequence which corresponds to the sequence which homology or identity is defined.
  • an antibody derived from a parent antibody which is defined by one or more amino acid sequences, such as specific CDR sequences or specific variable region sequences, in particular is an antibody having amino acid sequences, such as CDR sequences or variable region sequences, which are at least 75%, preferably at least 80%, at least 85%, at least 90%, at least 93%, at least 95%, at least 97%, at least 98% or at least 99% homologous or identical, especially identical, to the respective amino acid sequences of the parent antibody.
  • the antibody derived from (i.e. derivative of) a parent antibody comprises the same CDR sequences as the parent antibody, but differs in the remaining sequences of the variable regions.
  • antibody as used herein also refers to multivalent and multispecific antibodies, i.e. antibody constructs which have more than two binding sites each binding to the same epitope and antibody constructs which have one or more binding sites binding to a first epitope and one or more binding sites binding to a second epitope, and optionally even further binding sites binding to further epitopes.
  • Specific binding preferably means that an agent such as an antibody binds stronger to a target such as an epitope for which it is specific compared to the binding to another target.
  • An agent binds stronger to a first target compared to a second target if it binds to the first target with a dissociation constant (K d ) which is lower than the dissociation constant for the second target.
  • K d dissociation constant
  • the dissociation constant for the target to which the agent binds specifically is more than 100-fold, 200-fold, 500-fold or more than 1000-fold lower than the dissociation constant for the target to which the agent does not bind specifically.
  • the term “specific binding” in particular indicates a binding affinity between the binding partners with an affinity constant K a of at least 10 6 M ⁇ 1 , preferably at least 107 M ⁇ 1 , more preferably at least 10 8 M ⁇ 1 .
  • An antibody specific for a certain antigen in particular refers to an antibody which is capable of binding to said antigen with an affinity having a K a of at least 10 6 M ⁇ 1 , preferably at least 107 M ⁇ 1 , more preferably at least 10 8 M ⁇ 1 .
  • anti-MUC1 antibody in particular refers to an antibody specifically binding MUC1 and preferably is capable of binding to MUC1 with an affinity having a K a of at least 10 6 M ⁇ 1 , preferably at least 107 M ⁇ 1 , more preferably at least 10 8 M ⁇ 1 .
  • MUC1 refers to the protein MUC1, also known as mucin-1, polymorphic epithelial mucin (PEM) or cancer antigen 15-3, in particular to human MUC1 (Accession No. P15941).
  • MUC1 is a member of the mucin family and encodes a membrane bound, glycosylated phosphoprotein.
  • MUC1 has a core protein mass of 120-225 kDa which increases to 250-500 kDa with glycosylation. It extends 200-500 nm beyond the surface of the cell.
  • the protein is anchored to the apical surface of many epithelial cells by a transmembrane domain.
  • the extracellular domain includes a 20 amino acid variable number tandem repeat (VNTR) domain, with the number of repeats varying from 20 to 120 in different individuals. These repeats are rich in serine, threonine and proline residues which permits heavy O-glycosylation.
  • VNTR variable number tandem repeat
  • MUC1 refers to tumor-associated MUC1 (“TA-MUC1”). This MUC1 differs from MUC1 present on non-cancer cells in its much higher expression level, its localization and its glycosylation.
  • TA-MUC1 is present apolarly over the whole cell surface in cancer cells, while in non-cancer cells MUC1 has a strictly apical expression and hence, is not accessible for systemically administered antibodies. Furthermore, TA-MUC1 has an aberrant O-glycosylation which exposes new peptide epitopes on the MUC1 protein backbone and new carbohydrate tumor antigens such as the Thomsen-Friedenreich antigen alpha (TF ⁇ ).
  • TF ⁇ Thomsen-Friedenreich antigen alpha
  • TF ⁇ also called Thomsen-Friedenreich antigen alpha or Core-1, refers to the disaccharide Gal-ß1,3-GalNAc which is O-glycosidically linked in an alpha-anomeric configuration to the hydroxy amino acids serine or threonine of proteins in carcinoma cells.
  • sialic acid in particular refers to any N- or O-substituted derivatives of neuraminic acid. It may refer to both 5-N-acetylneuraminic acid and 5-N-glycolylneuraminic acid, but preferably only refers to 5-N-acetylneuraminic acid.
  • the sialic acid, in particular the 5-N-acetylneuraminic acid preferably is attached to a carbohydrate chain via a 2,3- or 2,6-linkage.
  • both 2,3- as well as 2,6-coupled sialic acids are present.
  • a “relative amount of glycans” according to the invention refers to a specific percentage or percentage range of the glycans attached to the antibodies of an antibody preparation or in a composition comprising antibodies, respectively.
  • the relative amount of glycans refers to a specific percentage or percentage range of all glycans comprised in the antibodies and thus, attached to the polypeptide chains of the antibodies in an antibody preparation or in a composition comprising antibodies.
  • 100% of the glycans refers to all glycans attached to the antibodies of the antibody preparation or in a composition comprising antibodies, respectively.
  • a relative amount of glycans carrying bisecting GlcNAc of 10% refers to a composition comprising antibodies wherein 10% of all glycans comprised in the antibodies and thus, attached to the antibody polypeptide chains in said composition comprise a bisecting GlcNAc residue while 90% of all glycans comprised in the antibodies and thus, attached to the antibody polypeptide chains in said composition do not comprise a bisecting GlcNAc residue.
  • the corresponding reference amount of glycans representing 100% may either be all glycan structures attached to the antibodies in the composition, or all N-glycans, i.e. all glycan structures attached to an asparagine residue of the antibodies in the composition, or all complex-type glycans.
  • the reference group of glycan structures generally is explicitly indicated or directly derivable from the circumstances by the skilled person.
  • N-glycosylation refers to all glycans attached to asparagine residues of the polypeptide chain of a protein. These asparagine residues generally are part of N-glycosylation sites having the amino acid sequence Asn-Xaa-Ser/Thr, wherein Xaa may be any amino acid except for proline. Likewise, “N-glycans” are glycans attached to asparagine residues of a polypeptide chain.
  • the terms “glycan”, “glycan structure”, “carbohydrate”, “carbohydrate chain” and “carbohydrate structure” are generally used synonymously herein.
  • N-glycans generally have a common core structure consisting of two N-acetylglucosamine (GlcNAc) residues and three mannose residues, having the structure Man ⁇ 1,6-(Man ⁇ 1,3-)Man ⁇ 1,4-GlcNAc ⁇ 1,4-GlcNAc ⁇ 1-Asn with Asn being the asparagine residue of the polypeptide chain.
  • N-glycans are subdivided into three different types, namely complex-type glycans, hybrid-type glycans and high mannose-type glycans.
  • the numbers given herein, in particular the relative amounts of a specific glycosylation property, are preferably to be understood as approximate numbers.
  • the numbers preferably may be up to 10% higher and/or lower, in particular up to 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2% or 1% higher and/or lower.
  • conjugate two or more compounds are linked together. In certain embodiments, at least some of the properties from each compound are retained in the conjugate. Linking may be achieved by a covalent or non-covalent bond. Preferably, the compounds of the conjugate are linked via a covalent bond.
  • the different compounds of a conjugate may be directly bound to each other via one or more covalent bonds between atoms of the compounds. Alternatively, the compounds may be bound to each other via a chemical moiety such as a linker molecule wherein the linker is covalently attached to atoms of the compounds. If the conjugate is composed of more than two compounds, then these compounds may, for example, be linked in a chain conformation, one compound attached to the next compound, or several compounds each may be attached to one central compound.
  • nucleic acid includes single-stranded and double-stranded nucleic acids and ribonucleic acids as well as deoxyribonucleic acids. It may comprise naturally occurring as well as synthetic nucleotides and can be naturally or synthetically modified, for example by methylation, 5′- and/or 3′-capping.
  • expression cassette in particular refers to a nucleic acid construct which is capable of enabling and regulating the expression of a coding nucleic acid sequence introduced therein.
  • An expression cassette may comprise promoters, ribosome binding sites, enhancers and other control elements which regulate transcription of a gene or translation of an mRNA.
  • the exact structure of expression cassette may vary as a function of the species or cell type, but generally comprises 5′-untranscribed and 5′- and 3′-untranslated sequences which are involved in initiation of transcription and translation, respectively, such as TATA box, capping sequence, CAAT sequence, and the like. More specifically, 5′-untranscribed expression control sequences comprise a promoter region which includes a promoter sequence for transcriptional control of the operatively connected nucleic acid. Expression cassettes may also comprise enhancer sequences or upstream activator sequences.
  • the term “promoter” refers to a nucleic acid sequence which is located upstream (5′) of the nucleic acid sequence which is to be expressed and controls expression of the sequence by providing a recognition and binding site for RNA-polymerases.
  • the “promoter” may include further recognition and binding sites for further factors which are involved in the regulation of transcription of a gene.
  • a promoter may control the transcription of a prokaryotic or eukaryotic gene.
  • a promoter may be “inducible”, i.e. initiate transcription in response to an inducing agent, or may be “constitutive” if transcription is not controlled by an inducing agent.
  • a gene which is under the control of an inducible promoter is not expressed or only expressed to a small extent if an inducing agent is absent. In the presence of the inducing agent the gene is switched on or the level of transcription is increased. This is mediated, in general, by binding of a specific transcription factor.
  • vector is used here in its most general meaning and comprises any intermediary vehicle for a nucleic acid which enables said nucleic acid, for example, to be introduced into prokaryotic and/or eukaryotic cells and, where appropriate, to be integrated into a genome.
  • Vectors of this kind are preferably replicated and/or expressed in the cells.
  • Vectors comprise plasmids, phagemids, bacteriophages or viral genomes.
  • plasmid as used herein generally relates to a construct of extrachromosomal genetic material, usually a circular DNA duplex, which can replicate independently of chromosomal DNA.
  • the term “host cell” relates to any cell which can be transformed or transfected with an exogenous nucleic acid.
  • the term “host cells” comprises according to the invention prokaryotic (e.g. E. coli ) or eukaryotic cells (e.g. mammalian cells, in particular human cells, yeast cells and insect cells). Particular preference is given to mammalian cells such as cells from humans, mice, hamsters, pigs, goats, or primates.
  • the cells may be derived from a multiplicity of tissue types and comprise primary cells and cell lines.
  • a nucleic acid may be present in the host cell in the form of a single copy or of two or more copies and, in one embodiment, is expressed in the host cell.
  • patient means according to the invention a human being, a nonhuman primate or another animal, in particular a mammal such as a cow, horse, pig, sheep, goat, dog, cat or a rodent such as a mouse and rat. In a particularly preferred embodiment, the patient is a human being.
  • cancer in particular comprises leukemias, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, endometrial cancer, kidney cancer, adrenal cancer, thyroid cancer, blood cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, liver cancer, colon cancer, stomach cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, pancreas cancer, ear, nose and throat (ENT) cancer, breast cancer, prostate cancer, bladder cancer, cancer of the uterus, ovarian cancer and lung cancer and the metastases thereof.
  • cancer according to the invention also comprises cancer metastases.
  • Tumor is meant a group of cells or tissue that is formed by misregulated cellular proliferation. Tumors may show partial or complete lack of structural organization and functional coordination with the normal tissue, and usually form a distinct mass of tissue, which may be either benign or malignant.
  • metastasis is meant the spread of cancer cells from its original site to another part of the body.
  • the formation of metastasis is a very complex process and normally involves detachment of cancer cells from a primary tumor, entering the body circulation and settling down to grow within normal tissues elsewhere in the body.
  • the new tumor is called a secondary or metastatic tumor, and its cells normally resemble those in the original tumor.
  • the secondary tumor is made up of abnormal breast cells, not of abnormal lung cells.
  • the tumor in the lung is then called metastatic breast cancer, not lung cancer.
  • composition particularly refers to a composition suitable for administering to a human or animal, i.e., a composition containing components which are pharmaceutically acceptable.
  • a pharmaceutical composition comprises an active compound or a salt or prodrug thereof together with a carrier, diluent or pharmaceutical excipient such as buffer, preservative and tonicity modifier.
  • the present invention is based on the development of a variant of the humanized anti-MUC1 antibody PankoMab wherein the glycosylation site in the CDR-H2 is deleted.
  • glycosylation site was achieved by substituting amino acid Asn (asparagine) 57 of the heavy chain variable region by another amino acid, especially Gln (glutamine). Asn 57 is the acceptor amino acid residue of the glycosylation site to which the carbohydrate structure is attached. Substituting this asparagine residue by another residue abolishes glycosylation because the carbohydrate structure can only be transferred to an asparagine residue by the enzymes of the host cell. It was surprisingly found that deletion of the glycosylation site in the CDR-H2 of PankoMab increased the antigen binding affinity of the antibody.
  • the present invention provides an antibody capable of binding to MUC1, which comprises
  • the antibody specifically binds to an epitope of MUC1.
  • “Specific binding” means binding which is not non-specific adsorption. Examples of criteria for determination on whether binding is specific or not can include a dissociation constant (herein referred to as “K”).
  • K dissociation constant
  • the epitope is in the extracellular tandem repeats of MUC1. In certain embodiments, the antibody binds to MUC1 in a glycosylation-dependent manner.
  • the antibody binds stronger if said tandem repeats are glycosylated at a threonine residue with N-acetyl galactosamine (Tn), sialyl ⁇ 2-6 N-acetyl galactosamine (sTn), galactose ß1-3 N-acetyl galactosamine (TF) or galactose ß1-3 (sialyl ⁇ 2-6)N-acetyl galactosamine (sTF), preferably with Tn or TF.
  • the carbohydrate moiety is bound to the threonine residue by an ⁇ -O-glycosidic bond.
  • the epitope in the tandem repeat domain of MUC1 in particular comprises the amino acid sequence PDTR (SEQ ID NO: 13) or PESR (SEQ ID NO: 14).
  • the binding to this epitope preferably is glycosylation dependent, as described above, wherein in particular the binding is increased if the carbohydrate moiety described above is attached to the threonine residue of the sequence PDTR or PESR (SEQ ID NOs: 13 and 14), respectively.
  • the epitope is a tumor-associated MUC1 epitope (TA-MUC1).
  • T-MUC1 epitope in particular refers to an epitope of MUC1 which is present on tumor cells but not on normal cells and/or which is only accessible by antibodies in the host's circulation when present on tumor cells but not when present on normal cells.
  • the binding of the antibody to cells expressing TA-MUC1 epitope is stronger than the binding to cells expressing normal, non-tumor MUC1.
  • said binding is at least 1.5-fold stronger, preferably at least 2-fold stronger, at least 5-fold stronger, at least 10-fold stronger or at least 100-fold stronger.
  • the antibody preferably specifically binds the glycosylated MUC1 tumor epitope such that the strength of the bond is increased at least by a factor 2, preferably a factor of 4 or a factor of 10, most preferably a factor of 20 in comparison with the bond to the non-glycosylated peptide of identical length and identical peptide sequence.
  • Said binding can be assayed or determined by ELISA, RIA, surface plasmon resonance (hereinafter, referred to as “SPR”) analysis, or the like.
  • Examples of equipment used in the SPR analysis can include BIAcoreTM (manufactured by GE Healthcare Bio-Sciences Crop.), ProteOnTM (manufactured by Bio-Rad Laboratories, Inc.), DRX2 Biosensor (manufactured by Dynamic Biosensors GmbH), SPR-NaviTM (manufactured by BioNavis Oy Ltd.), SpreetaTM (manufactured by Texas Instruments Inc.), SPRi-PlexllTM (manufactured by Horiba, Ltd.), and Autolab SPRTM (manufactured by Metrohm).
  • the binding of the antibody to the antigen expressed on cell surface can be assayed by flow cytometry or the like.
  • the antibody may exhibit antigen binding properties similar to those of a reference antibody comprising a heavy chain variable region with the amino acid sequence of SEQ ID NO: 11 or SEQ ID NO:10 and a light chain variable region with the amino acid sequence of SEQ ID NO: 12.
  • the reference antibody is the humanized antibody PankoMab.
  • the antibody according to the invention specifically binds to the same antigen as the reference antibody, and preferably binds to said antigen with a higher affinity. That is, the antibody preferably binds to the antigen with an affinity having a dissociation constant which is lower than that of the reference antibody, more preferably at least 10% lower, at least 20% lower, at least 30% lower or at least 50% lower.
  • the antibody preferably shows cross-specificity with the reference antibody comprising a heavy chain variable region with the amino acid sequence of SEQ ID NO: 11 and a light chain variable region with the amino acid sequence of SEQ ID NO: 12.
  • the humanized antibody is able to block the binding of the reference antibody to MUC1 if present in a high enough concentration. This is possible if the binding of the reference antibody to MUC1 is hindered when the antibody according to the invention is already bound to the antigen MUC1.
  • An antibody capable of binding to MUC1 of the present invention comprises a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • CDRs complementarity-determining regions
  • the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 9. Especially, the heavy chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 9. In these embodiments, the heavy chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 1, 2 and 3. Hence, any sequence deviations to SEQ ID NO: 9 are located in the framework regions, but not in the CDRs. In particular, the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 9.
  • CDR-H2 has the amino acid sequence of SEQ ID NO: 2, wherein the amino acid at position 8 of SEQ ID NO: 2 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine; especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine.
  • the amino acid at position 8 of SEQ ID NO: 2 is glutamine, histidine, tryptophan, lysine or arginine, especially glutamine.
  • CDR-H2 has the amino acid sequence of SEQ ID NO: 7.
  • the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 10.
  • the heavy chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 10.
  • the heavy chain variable region comprises the CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 7 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3.
  • any sequence deviations to SEQ ID NO: 10 are located in the framework regions, but not in the CDRs.
  • the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 10.
  • the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12. Especially, the light chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 12. In these embodiments, the light chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 4, 5 and 6. Hence, any sequence deviations to SEQ ID NO: 12 are located in the framework regions, but not in the CDRs. In particular, the light chain variable region comprises the amino acid sequence of SEQ ID NO: 12.
  • the heavy chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 9, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 2 and 3, and the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the heavy chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 9, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 2 and 3, and the light chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the heavy chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 10, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3, and the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the heavy chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 10, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3
  • the light chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20.
  • the heavy chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20.
  • the heavy chain variable region comprises the CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3.
  • any sequence deviations to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20 are located in the framework regions, but not in the CDRs.
  • the heavy chain variable region comprises the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20.
  • the amino acid at position 76 of SEQ ID NO: 20 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine; especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine.
  • the amino acid at position 76 of SEQ ID NO: 20 is glutamine, histidine, tryptophan, lysine or arginine, especially glutamine.
  • CDR-H2 has the amino acid sequence of SEQ ID NO: 7 and/or the heavy chain variable region comprises the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 23.
  • the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21.
  • the light chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21.
  • the light chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • any sequence deviations to amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21 are located in the framework regions, but not in the CDRs.
  • the light chain variable region comprises the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21.
  • the heavy chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3, and the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the heavy chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3
  • the light chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the heavy chain comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 15. Especially, the heavy chain comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 15.
  • the heavy chain comprises the CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3.
  • any sequence deviations to SEQ ID NO: 15 are located in the framework regions, but not in the CDRs.
  • the heavy chain comprises the amino acid sequence of SEQ ID NO: 15.
  • the amino acid at position 57 of SEQ ID NO: 15 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine; especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine.
  • the amino acid at position 57 of SEQ ID NO: 15 is glutamine, histidine, tryptophan, lysine or arginine, especially glutamine.
  • CDR-H2 has the amino acid sequence of SEQ ID NO: 7 and/or the heavy chain variable region comprises the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 22.
  • the light chain comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 16. Especially, the light chain comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 16. In these embodiments, the light chain still comprises CDRs having the amino acid sequences of SEQ ID NOs: 4, 5 and 6. Hence, any sequence deviations to SEQ ID NO: 16 are located in the framework regions, but not in the CDRs. In particular, the light chain comprises the amino acid sequence of SEQ ID NO: 16.
  • the heavy chain has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 15, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3
  • the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 16, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the heavy chain has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 15, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3
  • the light chain has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 16, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the antibody according to the present invention includes and encompasses modified forms thereof.
  • the modified form of the antibody of the present invention means an antibody of the present invention provided with chemical or biological modification.
  • the chemically modified form includes forms wherein the amino acid skeleton is conjugated with a chemical moiety, forms having a chemically modified N-linked or O-linked carbohydrate chain, and the like. Said chemical moiety or form can be toxic or cytotoxic.
  • Biologically modified forms include forms that have undergone post-translational modification (e.g., N-linked or O-linked glycosylation, N-terminal or C-terminal processing, deamidation, isomerization of aspartic acid, or oxidation of methionine), forms containing a methionine residue added to the N-terminus by expression using prokaryotic host cells, and the like.
  • a modified form is also meant to include a form labeled to permit detection or isolation of the antibody of the present invention or the antigen thereof, for example, enzyme-labeled forms, fluorescently labeled forms, and affinity-labeled forms.
  • Such a modified form of the antibody of the present invention is useful in improvement in the stability or blood retention of the original antibody of the present invention, reduction in antigenicity, detection or isolation of the antibody or the antigen, etc.
  • the antibody may comprise one or more modifications selected from the group consisting of defucosylation, reduced fucose, N-linked glycosylation, O-linked glycosylation, N-terminal processing, C-terminal processing, deamidation, isomerization of aspartic acid, oxidation of methionine, substitutions of two leucine (L) residues to alanine (A) at position 234 and 235 of the heavy chain (LALA), amidation of a proline residue and deletion or lack of one, two, or three amino acids at the carboxyl terminus.
  • modifications selected from the group consisting of defucosylation, reduced fucose, N-linked glycosylation, O-linked glycosylation, N-terminal processing, C-terminal processing, deamidation, isomerization of aspartic acid, oxidation of methionine, substitutions of two leucine (L) residues to alanine (A) at position 234 and 235 of the heavy chain (LALA), amidation
  • the antibody lacks one, two, or three carboxyl-terminal amino acid(s) at one or both heavy chains, or it lacks one carboxyl-terminal amino acid and the carboxyl-terminal proline residues is amidated at one or both heavy chains.
  • Such a modification may be made at an arbitrary position or the desired position in the antibody thereof.
  • the same or two or more different modifications may be made at one or two or more positions therein.
  • antibodies produced by cultured mammalian cells are known to lack a carboxyl-terminal lysine residue in its heavy chain (Journal of Chromatography A, 705: 129-134 (1995)). It is also known that occasionally 2 carboxyl-terminal amino acid residues (i.e., glycine and lysine) of a heavy chain are missing and that a proline residue newly located at the carboxyl terminus is amidated (Analytical Biochemistry, 360: 75-83 (2007)). Such lack or modification in these heavy chain sequences, however, affects neither the ability of the antibody to bind to its antigen nor the effector functions (complement activation, antibody-dependent cytotoxicity, etc.) of the antibody.
  • the antibody comprises a deletion or lack of 1 or 2 amino acid(s) in the carboxyl terminus of the heavy chain, and has an amidated residue (e.g., an amidated proline residue at the carboxyl-terminal site of the heavy chain).
  • an amidated residue e.g., an amidated proline residue at the carboxyl-terminal site of the heavy chain.
  • the antibody according to the present invention is not limited to the types described above as long as the deletion mutant maintains the ability to bind to the antigen.
  • two heavy chains of the antibody according to the present invention may be composed of any one type of heavy chain selected from the group consisting of the full length heavy chains and the heavy chains of the deletion mutant or may be composed of the combination of any two types selected therefrom.
  • the quantitative ratio of the deletion variant heavy chain(s) depends on the type of cultured mammalian cells producing the antibody according to the present invention, and the culture conditions of the cells.
  • the antibody according to the present invention can include two heavy chains, both of which lack one carboxyl-terminal amino acid residue.
  • the antibody comprises the heavy chain having an amino acid sequence represented by amino acid Nos 1 to 446 of SEQ ID NO: 15 or 22 and the light chain having an amino acid sequence represented by amino acid Nos 1 to 219 of SEQ ID NO: 16.
  • the amino acid at position 57 of SEQ ID NO: 15 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine; especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine.
  • the amino acid at position 57 of SEQ ID NO: 15 is glutamine, histidine, tryptophan, lysine or arginine, especially glutamine.
  • the antibody according to the present invention competes for the binding to TA-MUC1 with an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region having the amino acid sequence of SEQ ID NO: 12, or an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 11 and a light chain variable region having the amino acid sequence of SEQ ID NO: 12.
  • the antibody has the following properties: (a) specifically binding to MUC1, and/or (b) having the activity of being internalized into MUC1-expressing cells through binding to MUC1.
  • the antibody comprises at least one antibody heavy chain.
  • the antibody comprises two antibody heavy chains.
  • the antibody heavy chains in particular comprise a VH domain, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain.
  • the antibody heavy chains comprise a CH2 domain and a CH3 domain, but do not comprise a CH1 domain.
  • one or more constant domains of the heavy chains may be replaced by other domains, in particular similar domains such as for example albumin.
  • the antibody heavy chains may be of any type, including ⁇ -, ⁇ -, ⁇ -, ⁇ - and ⁇ -chains, and preferably are ⁇ -chains, including ⁇ 1-, ⁇ 2-, ⁇ 3- and ⁇ 4-chains, especially ⁇ 1-chains.
  • the antibody preferably is an IgG-type antibody such as an IgG1-, IgG3- or IgG4-type antibody, in particular an IgG1-type antibody.
  • the antibody further comprises at least one antibody light chain, especially two antibody light chains.
  • the antibody light chains in particular comprise a VL domain and a CL domain.
  • the antibody light chain may be a ⁇ -chain or a ⁇ -chain and especially is a ⁇ -chain.
  • the antibody comprises two antibody heavy chains and two antibody light chains.
  • the antibody comprises two antibody heavy chains of the ⁇ 1-type, each comprising a VH domain, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain, and two antibody light chains of the ⁇ -type, each comprising a VL domain and a CL domain.
  • the antibody does not comprise an antibody light chain.
  • the light chain variable region may be fused to the N terminus of the heavy chain variable region or is inserted C terminal to the heavy chain variable region.
  • Peptide linkers may be present to connect the light chain variable region with the remaining parts of the heavy chain.
  • the antibody comprises an Fc region.
  • the antibody may especially be a whole antibody, comprising two heavy chains each comprising the domains VH, CH1, hinge region, CH2 and CH3, and two light chains each comprising the domains VL and CL.
  • the antibody in particular is capable of binding to one or more human Fc ⁇ receptors, especially human Fc ⁇ receptor IIIA.
  • the antibody does not or not significantly bind the human Fc ⁇ receptor IIIA, and especially does not or not significantly bind to any human Fc ⁇ receptor.
  • the antibody in particular does not comprise a glycosylation site in the CH2 domain.
  • the antibody does not comprise an Fc region.
  • the antibody in particular is a single chain variable region fragment (scFv) or another antibody fragment not comprising an Fc region.
  • the anti-MUC1 antibody may comprise a CH2 domain in one or more antibody heavy chains.
  • Natural human antibodies of the IgG type comprise an N-glycosylation site in the CH2 domain.
  • the CH2 domains present in the antibody may or may not comprise an N-glycosylation site.
  • the antibody does not comprise a glycosylation site in the CH2 domain.
  • the antibody does not comprise an asparagine residue at the position in the heavy chain corresponding to position 297 according to the IMGT/Eu numbering system.
  • the antibody may comprise an Ala297 mutation in the heavy chain.
  • the antibody preferably has a strongly reduced ability or completely lacks the ability to induce, via binding to Fc ⁇ receptors, antibody-dependent cellular cytotoxicity (ADCC) and/or antibody-dependent cellular phagocytosis (ADCP) and/or complement-dependent cytotoxicity (CDC).
  • Strongly reduced ability in this respect in particular refers to a reduction to 10% or less, especially 3% or less, 1% or less or 0.1% or less activity compared to the same antibody comprising an N-glycosylation site in its CH2 domains and having a common mammalian glycosylation pattern such as those obtainable by production in human cell lines or in CHO cell lines, for example a glycosylation pattern as described herein.
  • the antibody in particular is an IgG1-type antibody.
  • the CH2 domains present in the antibody comprise an N-glycosylation site.
  • This glycosylation site in particular is at an amino acid position corresponding to amino acid position 297 of the heavy chain according to the IMGT/Eu numbering system and has the amino acid sequence motive Asn Xaa Ser/Thr wherein Xaa may be any amino acid except proline.
  • the N-linked glycosylation at Asn297 is conserved in mammalian IgGs as well as in homologous regions of other antibody isotypes. Due to optional additional amino acids which may be present in the variable region or other sequence modifications, the actual position of this conserved glycosylation site may vary in the amino acid sequence of the antibody.
  • the glycans attached to the antibody are biantennary complex type N-linked carbohydrate structures, preferably comprising at least the following structure:
  • Asn is the asparagine residue of the polypeptide portion of the antibody; GlcNAc is N-acetylglucosamine and Man is mannose.
  • the terminal GlcNAc residues may further carry a galactose residue, which optionally may carry a sialic acid residue.
  • a further GlcNAc residue (named bisecting GlcNAc) may be attached to the Man nearest to the polypeptide.
  • a fucose may be bound to the GlcNAc attached to the Asn.
  • the antibody in particular is an IgG1-type antibody.
  • the antibody does not comprise N-glycolyl neuraminic acids (NeuGc) or detectable amounts of NeuGc.
  • the antibody preferably also does not comprise Galili epitopes (Gal ⁇ 1,3-Gal structures) or detectable amounts of the Galili epitope.
  • the relative amount of glycans carrying NeuGc and/or Gal ⁇ 1,3-Gal structures is less than 0.1% or even less than 0.02% of the total amount of glycans attached to the CH2 domains of the antibodies in the population of antibodies.
  • the antibody has a human glycosylation pattern. Due to these glycosylation properties, foreign immunogenic non-human structures which induce side effects are absent which means that unwanted side effects or disadvantages known to be caused by certain foreign sugar structures such as the immunogenic non-human sialic acids (NeuGc) or the Galili epitope (Gal-Gal structures), both known for rodent production systems, or other structures like immunogenic high-mannose structures as known from e.g. yeast systems are avoided.
  • the immunogenic non-human sialic acids NeGc
  • Gal-Gal structures Galili epitope
  • the antibody comprises a glycosylation pattern having a detectable amount of glycans carrying a bisecting GlcNAc residue.
  • the relative amount of glycans carrying a bisecting GlcNAc residue is at least 0.5%, especially at least 1% of the total amount of glycans attached to the glycosylation sites of the antibody in a composition.
  • the glycosylation pattern comprises a relative amount of glycans carrying at least one galactose residue of at least 25% of the total amount of glycans attached to the antibody in a composition.
  • the relative amount of glycans carrying at least one galactose residue is at least 30%, especially at least 35% or at least 40% of the total amount of glycans attached to the antibody in a composition.
  • the glycosylation pattern comprises a relative amount of glycans carrying at least one sialic acid residue of at least 1% of the total amount of glycans attached to the antibody in a composition.
  • the relative amount of glycans carrying at least one sialic acid residue is at least 1.5%, especially at least 2% of the total amount of glycans attached to the antibody in a composition.
  • the antibody may have a glycosylation pattern having a high amount of core fucose or a low amount of core fucose. A reduced amount of fucosylation increases the ability of the antibody to induce ADCC.
  • the relative amount of glycans carrying a core fucose residue is 40% or less, especially 30% or less or 20% or less of the total amount of glycans attached to the antibody in a composition.
  • the relative amount of glycans carrying a core fucose residue is at least 60%, especially at least 65% or at least 70% of the total amount of glycans attached to the antibody in a composition.
  • the ability of the antibody to induce ADCC and the strength of said ADCC induction can be controlled.
  • the ADCC activity is increased by glycosylation of the Fc part of the antibody and further by reducing the amount of fucosylation in said glycosylation. In certain applications, fine tuning of the ADCC activity is important.
  • the antibody without a glycosylation site in the CH2 domain, the antibody with a glycosylation site in the CH2 domain and with a high amount of fucosylation, or the antibody with a glycosylation site in the CH2 domain and with a low amount of fucosylation may be most advantageous.
  • the antibody is preferably recombinantly produced in a host cell.
  • the host cell used for the production of the antibody may be any host cells which can be used for antibody production. Suitable host cells are in particular eukaryotic host cells, especially mammalian host cells. Exemplary host cells include yeast cells such as Pichia pastoris cell lines, insect cells such as SF9 and SF21 cell lines, plant cells, bird cells such as EB66 duck cell lines, rodent cells such as CHO, NS0, SP2/0 and YB2/0 cell lines, and human cells such as HEK293, PER.C6, CAP, CAP-T, AGE1.HN, Mutz-3 and KG1 cell lines.
  • the antibody is produced recombinantly in a human blood cell line, in particular in a human myeloid leukemia cell line.
  • a human myeloid leukemia cell line Preferred human cell lines which can be used for production of the antibody as well as suitable production procedures are described in WO 2008/028686 A2.
  • the antibody is obtained by expression in a human myeloid leukemia cell line selected from the group consisting of NM-H9D8, NM-H9D8-E6 and NM-H9D8-E6Q12 and cell lines derived therefrom. These cell lines were deposited under the accession numbers DSM ACC2806 (NM-H9D8; deposited on Sep.
  • NM-H9D8 cells provide a glycosylation pattern with a high degree of sialylation, a high degree of bisecting GlycNAc, a high degree of galactosylation and a high degree of fucosylation.
  • NM-H9D8-E6 and NM-H9D8-E6Q12 cells provide a glycosylation pattern similar to that of NM-H9D8 cells, except that the degree of fucosylation is very low.
  • suitable cell lines include K562, a human myeloid leukemia cell line present in the American Type Culture Collection (ATCC CCL-243), as well as cell lines derived from the aforementioned.
  • the antibody is produced recombinantly in CHO cells.
  • the antibody may be produced recombinantly in a CHO dhfr- cell line such as the cell line of ATCC No. CRL-9096.
  • the antibody comprises one or more further agents conjugated thereto.
  • the further agent may be any agent suitable for conjugation to the antibody. If more than one further agent is present in the antibody, these further agents may be identical or different, and in particular are all identical. Conjugation of the further agent to the antibody can be achieved using any methods known in the art.
  • the further agent may be covalently, in particular by fusion or chemical coupling, or non-covalently attached to the antibody.
  • the further agent is covalently attached to the antibody, especially via a linker moiety.
  • the linker moiety may be any chemical entity suitable for attaching the further agent to the antibody.
  • the further agent preferably is useful in therapy, diagnosis, prognosis, detecting and/or monitoring of a disease, in particular cancer.
  • the further agent may be selected from the group consisting of radionuclides, chemotherapeutic agents, antibodies or antibody fragments, in particular those of different specificity than the anti-MUC1 antibody, e.g. checkpoint antibodies which block or activate immunomodulatory targets, enzymes, interaction domains, detectable labels, toxins, cytolytic components, immunomodulators, immunoeffectors, MHC class I or class II antigens, and liposomes.
  • a particular preferred further agent is a radionuclide or a cytotoxic agent capable of killing cancer cells, such as a chemotherapeutic agent.
  • a chemotherapeutic agent is attached to the anti-MUC1 antibody forming a conjugate.
  • alkylating agents such as cisplatin, anti-metabolites, plant alkaloids and terpenoids, vinca alkaloids, podophyllotoxin, taxanes such as taxol, topoisomerase inhibitors such as irinotecan and topotecan, antineoplastics such as doxorubicin or microtubule inhibitors such as auristatins and maytansin/maytansinoids.
  • the chemotherapeutic agent may in particular be selected from a group consisting of a V-ATPase inhibitor, a pro-apoptotic agent, a Bcl2 inhibitor, an MCL1 inhibitor, a HSP90 inhibitor, an IAP inhibitor, an mTor inhibitor, a microtubule stabilizer, a microtubule destabilizer, an auristatin, a dolastatin, a maytansin, a maytansinoid, amatoxin, a methionine aminopeptidase, an inhibitor of nuclear export of proteins CRM1, a DPPIV inhibitor, proteasome inhibitors, inhibitors of phosphoryl transfer reactions in mitochondria, a protein synthesis inhibitor, a kinase inhibitor, a CDK2 inhibitor, a CDK9 inhibitor, a kinesin inhibitor, an HDAC inhibitor, a DNA damaging agent, a DNA alkylating agent, a DNA intercalator, a DNA minor groove binder, a DHFR inhibitor, an inhibitor of
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is selected from the group consisting of an auristatin, a microtubule inhibitor such as maytansinoid, a DNA damaging agent, a DNA alkylating agent and a DNA minor groove binder.
  • chemotherapeutic agent is a maytansin or maytansinoid.
  • maytansinoids useful for conjugation include maytansinol, N 2′ -deacetyl-N 2′ -(3-mercapto-1-oxopropyl)-maytansine (DM1), N 2′ -deacetyl-N 2′ -(4-mercapto-1-oxopentyl)-maytansine (DM3), and N 2′ -deacetyl-N 2′ -(4-methyl-4-mercapto-1-oxopentyl)-maytansine (DM4).
  • DM1 or DM4 is attached to the anti-MUC1 antibody.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is an auristatin, in particular monomethyl auristatin F (MMAF), monomethyl auristatin E (MMAE) or auristatin T.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is a DNA minor groove binder, in particular pyrrolobenzodiazepine (PBD), pyrrolobenzodiazepine dimer (PBD dimer), duocarmycin, duocarmycin-hydroxybenzamide-azaindole (DUBA), seco-duocarmycin-hydroxybenzamide-azaindole (seco-DUBA) or doxorubicin.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is a DNA alkylating agent, in particular indolinobenzodiazepine or oxazolidinobenzodiazepine.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is a DNA damaging agent, in particular calicheamicin.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is an inhibitor of microtubule formation, in particular a tubulysin, an ansamitocin, podophyllotoxin or vinblastine.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is a stabilizer of microtubuli, in particular paclitaxel or an epothilone. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a stabilizer of actin, in particular a phallotoxin. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a topoisomerase II inhibitor, in particular teniposide, XK469, razoxane, amsacrine, idarubicin or mebarone.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is a platinum compound, in particular cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin or sattraplatin.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is a ribosome inhibitor, in particular ricin, saporin, abrin, diphtheria toxin or exotoxin A.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is an RNA polymerase II inhibitor, in particular an amatoxin, such as, for example, amanitin.
  • the chemotherapeutic agent attached to the anti-MUC1 antibody is a bacterial toxin, in particular anthrax toxin.
  • Suitable antibody drug conjugates are also described in EP 16 151 774.3 and LU 92659, to which is explicitly referred to herewith.
  • the further agent is a polypeptide or protein. This polypeptide or protein may in particular be fused to a polypeptide chain of the antibody. In certain embodiments, the further agent being a polypeptide or protein is fused to the C terminus of an antibody light chain of the antibody. In embodiments wherein the antibody comprises two antibody light chains, a further agent being a polypeptide or protein may be fused to the C terminus of each of the two antibody light chains. In further embodiments, the further agent being a polypeptide or protein is fused to the C terminus of an antibody heavy chain of the antibody. In embodiments wherein the antibody comprises two antibody heavy chains, a further agent being a polypeptide or protein may be fused to the C terminus of each of the two antibody heavy chains.
  • the further agents may be identical or different and in particular have the same amino acid sequence.
  • Suitable examples of such further agents being a polypeptide or protein may be selected from the group consisting of cytokines, chemokines, antibodies, antigen binding fragments, enzymes, and interaction domains.
  • the further agent being a polypeptide or protein is a checkpoint antibody which blocks and/or triggers activating signals.
  • targets include CD40, CD3, CD137 (4-1BB), OX40, GITR, CD27, CD278 (ICOS), CD154 (CD40 ligand), CD270 (HVEM) and CD258 (LIGHT) as activating targets, CTLA4, PD1, CD80, CD244, A2AR, B7-H3 (CD276), B7-H4 (VTCN1), BTLA, IDO, KIR, LAG3, TIM-3, VISTA and phosphatidylserine as inhibitory targets, and their respective ligands such as PDL1.
  • the anti-MUC1 antibody comprises two heavy chains and two light chains as described herein, wherein a scFv fragment specifically binding to CD3 is fused to the C terminus of each heavy chain; or wherein a scFv fragment specifically binding to PDL1 is fused to the C terminus of each light chain.
  • the further agent being a polypeptide or protein is an immunomodulatory compound such as a chemokine, cytokine or growth factor.
  • cytokines in this respect include interferons such as interferon-a, interferon-p and interferon-y, and interleukins.
  • Suitable growth factors include G-CSF and GM-CSF.
  • the present invention provides a nucleic acid encoding the antibody.
  • the nucleic acid sequence of said nucleic acid may have any nucleotide sequence suitable for encoding the antibody. However, preferably the nucleic acid sequence is at least partially adapted to the specific codon usage of the host cell or organism in which the nucleic acid is to be expressed, in particular the human codon usage.
  • the nucleic acid may be double-stranded or single-stranded DNA or RNA, preferably double-stranded DNA such as cDNA or single-stranded RNA such as mRNA. It may be one consecutive nucleic acid molecule or it may be composed of several nucleic acid molecules, each coding for a different part of the antibody.
  • the present invention provides a nucleotide sequence of the heavy chain of PankoMab variant (PM-N54Q) represented by SEQ ID NO: 17 and a nucleotide sequence of the light chain of PankoMab variant (PM-N54Q) represented by SEQ ID NO: 18.
  • the nucleic acid may, for example, be a single nucleic acid molecule containing several coding regions each coding for one of the amino acid chains of the antibody, preferably separated by regulatory elements such as IRES elements in order to generate separate amino acid chains, or the nucleic acid may be composed of several nucleic acid molecules wherein each nucleic acid molecule comprises one or more coding regions each coding for one of the amino acid chains of the antibody.
  • the nucleic acid may also comprise further nucleic acid sequences or other modifications which, for example, may code for other proteins, may influence the transcription and/or translation of the coding region(s), may influence the stability or other physical or chemical properties of the nucleic acid, or may have no function at all.
  • the present invention provides an expression cassette or vector comprising a nucleic acid according to the invention and a promoter operatively connected with said nucleic acid.
  • the expression cassette or vector may comprise further elements, in particular elements which are capable of influencing and/or regulating the transcription and/or translation of the nucleic acid, the amplification and/or reproduction of the expression cassette or vector, the integration of the expression cassette or vector into the genome of a host cell, and/or the copy number of the expression cassette or vector in a host cell.
  • Suitable expression cassettes and vectors comprising respective expression cassettes for expressing antibodies are well known in the prior art and thus, need no further description here.
  • the present invention provides a host cell comprising the nucleic acid according to the invention or the expression cassette or vector according to the invention.
  • the host cell may be any host cell. It may be an isolated cell or a cell comprised in a tissue.
  • the host cell is a cultured cell, in particular a primary cell or a cell of an established cell line, preferably a tumor-derived cell.
  • it is a bacterial cell such as E. coli , a yeast cell such as a Saccharomyces cell, in particular S.
  • the host cell is derived from human myeloid leukaemia cells.
  • it is selected from the following cells or cell lines: K562, KG1, MUTZ-3, or a cell or cell line derived therefrom, or a mixture of cells or cell lines comprising at least one of those aforementioned cells.
  • the host cell is preferably selected from the group consisting of NM-H9D8, NM-H9D8-E6, NM H9D8-E6012, and a cell or cell line derived from anyone of said host cells. These cell lines and their properties are described in detail in the PCT-application WO 2008/028686 A2.
  • the host cell is of a CHO dhfr- cell line such as the cell line of ATCC No. CRL-9096.
  • the host cell is optimized for expression of glycoproteins, in particular antibodies, having a specific glycosylation pattern.
  • the codon usage in the coding region of the nucleic acid according to the invention and/or the promoter and the further elements of the expression cassette or vector are compatible with and, more preferably, optimized for the type of host cell used.
  • the antibody is produced by a host cell or cell line as described above.
  • the present invention provides a method of producing the antibody in a host cells as described herein.
  • the method in particular comprises the steps of providing a host cell comprising a nucleic acid encoding the antibody, culturing the host cell under conditions suitable for expression of the antibody, and obtaining the antibody expressed by the host cell.
  • the antibody according to the invention may be obtained or obtainable by said method.
  • the present invention provides a composition comprising the antibody, the nucleic acid, the expression cassette or vector, or the host cell.
  • the composition may also contain more than one of these components.
  • the composition may comprise one or more further components selected from the group consisting of solvents, diluents, and excipients
  • the composition is a pharmaceutical composition.
  • the components of the composition preferably are all pharmaceutically acceptable.
  • the composition may be a solid or fluid composition, in particular a—preferably aqueous—solution, emulsion or suspension or a lyophilized powder.
  • the antibody in particular is useful in medicine, in particular in therapy, diagnosis, prognosis, detecting and/or monitoring of a disease, in particular a disease as described herein, preferably cancer, infections, inflammatory diseases, graft-versus-host disease and immunodeficiencies.
  • the invention provides the antibody, the nucleic acid, the expression cassette or vector, the host cell, or the composition for use in medicine.
  • the use in medicine is a use in the treatment, prognosis, diagnosis, detecting and/or monitoring of a disease such as, for example, diseases associated with abnormal cell growth such as cancer, infections such as bacterial, viral, fungal or parasitic infections, inflammatory diseases such as autoimmune diseases and inflammatory bowel diseases, and diseases associated with a reduce immune activity such as immunodeficiencies.
  • a disease such as, for example, diseases associated with abnormal cell growth such as cancer, infections such as bacterial, viral, fungal or parasitic infections, inflammatory diseases such as autoimmune diseases and inflammatory bowel diseases, and diseases associated with a reduce immune activity such as immunodeficiencies.
  • the disease is cancer.
  • the cancer has a detectable expression of MUC1 or TA-MUC1, preferably detectable by immunohistochemistry, ELISA, RIA, enzyme-linked immunospot (ELISPOT) assay, dot blotting, Ouchterlony test or counter-immunoelectrophoresis (CIE), or in-situ hybridization. It especially includes cells having an MUC1 or TA-MUC1 expression which is detectable by immunohistochemistry or in-situ hybridization.
  • the cancer may be tested on MUC1 or TA-MUC1 level prior to administration of the anti-MUC1 antibody.
  • kits and devices comprising the antibody according to the invention, and associated methods that are useful in the diagnosis, detecting or monitoring of MUC1 associated disorders such as cancer.
  • a sandwich ELISA kit for testing, detecting or diagnosis comprising the antibody of the present invention is provided.
  • This kit may further comprise one or more of a solution of MUC1 or TA-MUC1 protein standards, a coloring reagent, a buffer solution for dilution, an antibody for solid phase, an antibody for detection, and a washing solution, and the like.
  • the amount of the antibody bound to the antigen can be measured by the application of a method such as an absorbance, fluorescence, luminescence, or radioisotope (RI) method.
  • RI radioisotope
  • an absorbance plate reader, a fluorescence plate reader, a luminescence plate reader, an RI liquid scintillation counter, or the like is used in the measurement.
  • the present invention provides the antibody according to the invention for use in immunohistochemistry (IHC) analysis, and a composition comprising the same.
  • IHC immunohistochemistry
  • the immunohistochemistry is not particularly limited as long as this approach involves reacting a tissue section with an antigen-binding antibody (primary antibody) and detecting the primary antibody bound with the antigen.
  • an antigen-binding antibody primary antibody
  • the cancer can in particular be selected from the group consisting of colon cancer, lung cancer, ovarian cancer, breast cancer such as triple negative breast cancer, pancreatic cancer, cervical cancer, endometrial cancer, gastrointestinal cancer, kidney cancer, head and neck cancer, thyroid cancer and urothelial cancer.
  • the cancer may further in particular be selected from stomach cancer, liver cancer, bladder cancer, skin cancer, prostate cancer and blood cancer.
  • the cancer is a metastasizing cancer.
  • the cancer may include any type of metastases, such as skin metastases, lymph node metastases, lung metastases, liver metastases, peritoneal metastases, pleural metastases and/or brain metastases.
  • the cancer has an inflammatory phenotype.
  • any of the cancer types described above may be an inflammatory cancer.
  • the viral infection is caused by human immunodeficiency virus, herpes simplex virus, Epstein Barr virus, influenza virus, lymphocytic choriomeningitis virus, hepatitis B virus or hepatitis C virus.
  • the inflammatory disease may be selected from inflammatory bowel disease, pelvic inflammatory disease, ischemic stroke, Alzheimer's disease, asthma, pemphigus vulgaris and dermatitis/eczema.
  • the autoimmune disease may be selected from the group consisting of celiac disease, diabetes mellitus type 1, Graves' disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis, systemic lupus erythematosus, vitiligo, psoriatic arthritis, atopic dermatitis, scleroderma, sarcoidosis, primary biliary cirrhosis, Guillain-Barre syndrome, autoimmune hepatitis and ankylosing spondylitis.
  • the disease comprises or is associated with cells which express MUC1, especially TA-MUC1.
  • a cancer to be treated is MUC1 positive, especially TA-MUC1 positive, i.e. comprises cancer cells which express MUC1, especially TA-MUC1.
  • the antibody is used for treatment in combination with another therapeutic agent, especially for treatment of cancer in combination with another anti-cancer agent.
  • Said further therapeutic agent may be any known anti-cancer agent.
  • Suitable anti-cancer therapeutic agents which may be combined with the antibody according to the invention may be chemotherapeutic agents, other antibodies, immunostimulatory agents, cytokines, chemokines, and vaccines.
  • therapy with the antibody may be combined with radiation therapy, surgery and/or traditional Chinese medicine.
  • Anti-cancer agents that can be used in combination with the anti-MUC1 antibody may be selected from any chemotherapeutic agent, in particular chemotherapeutic agents known to be effective for treatment of MUC1 positive cancers.
  • the type of chemotherapeutic agent also depends on the cancer to be treated.
  • the combination partner may be selected from the group consisting of taxanes such as paclitaxel (Taxol), docetaxel (Taxotere) and SB-T-1214; cyclophosphamide; imatinib; pazopanib; capecitabine; cytarabine; vinorelbine; gemcitabine; anthracyclines such as daunorubicin, doxorubicin, epirubicin, idarubicin, valrubicin and mitoxantrone; aromatase inhibitors such as aminoglutethimide, testolactone (Teslac), anastrozole (Arimidex), letrozole (Femara), exemestane (Aromasin), vorozole (Rivizor), formestane (Lentaron), fadrozole (Afema), 4-hydroxyandrostenedione, 1,4,6-androstatrien-3,17-dione (
  • therapeutic antibodies can be used as further combination partner. It may be any antibody that is useful in cancer therapy which is different from the anti-MUC1 antibody.
  • the further antibody is approved for cancer treatment by an administration such as the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA, formerly EMEA) and the Bundesinstitut for Arzneistoff und Kunststoff app (BfArM).
  • anti-EGFR antibodies such as Cetuximab, Tomuzotuximab, Panitumumab, Zalutumumab, Nimotuzumab, Matuzumab and Necitumumab
  • anti-HER2 antibodies such as Trastuzumab, Timigutuzumab and Pertuzumab
  • anti-VEGF antibodies such as bevacizumab (Avastin)
  • anti-CD52 antibodies such as alemtuzumab (Campath)
  • anti-CD30 antibodies such as brentuximab (Adcetris)
  • anti-CD33 antibodies such as gemtuzumab (Mylotarg)
  • anti-CD20 antibodies such as rituximab (Rituxan, Mabthera), tositumomab (Bexxar) and ibritumomab (Zevalin).
  • antibodies suitable for combination with the cancer therapy described herein include antibodies against antigens selected from the group consisting of Thomsen-Friedenreich antigen (TF ⁇ , TFP), Tn, Lewis Y, CD44, folate receptor a, NeuGc-GM3 ganglioside, DLL-3, RANKL, PTK7, Notch-3, Ephrin A4, insulin-like growth factor receptor 1, activin receptor-like kinase-1, claudin-6, disialoganglioside GD2, endoglin, transmembrane glycoprotein NMB, CD56, tumor-associated calcium signal transducer 2, tissue factor, ectonucleotide pyrophosphatase/phosphodiesterase 3, CD70, P-cadherin, mesothelin, six transmembrane epithelial antigen of the prostate 1 (STEAP1), carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), nectin 4, guanylyl
  • the anti-MUC1 antibody can further be combined with checkpoint antibodies, i.e. antibodies blocking or activating immunomodulatory targets. Thereby, inhibitory signals for an immune response can be blocked and/or activating signals can be triggered.
  • checkpoint antibodies i.e. antibodies blocking or activating immunomodulatory targets.
  • inhibitory signals for an immune response can be blocked and/or activating signals can be triggered.
  • targets include CD40, CD3, CD137 (4-1BB), OX40, GITR, CD27, CD278 (ICOS), CD154 (CD40 ligand), CD270 (HVEM) and CD258 (LIGHT) as activating targets, CTLA4, PD1, CD80, CD244, A2AR, B7-H3 (CD276), B7-H4 (VTCN1), BTLA, IDO, KIR, LAG3, TIM-3, VISTA and phosphatidylserine as inhibitory targets, and their respective ligands such as PDL1.
  • the anti-MUC1 antibody can be combined with the treatment with immunomodulatory compounds such as chemokines, cytokines, growth factors and vaccines.
  • immunomodulatory compounds such as chemokines, cytokines, growth factors and vaccines.
  • Suitable cytokines in this respect include interferons such as interferon- ⁇ , interferon- ⁇ and interferon- ⁇ , and interleukins.
  • Suitable growth factors include G-CSF and GM-CSF.
  • the anti-MUC1 antibody preferably is used for treatment of a primary tumor, a recurrent tumor and/or metastases of such tumors, and in particular is used for treatment before, during or after surgery and for the prevention or treatment of metastases.
  • the anti-MUC1 antibody in particular is for the treatment of a patient as adjuvant therapy.
  • the anti-MUC1 antibody is for the treatment of a patient as neoadjuvant therapy or in a combined neoadjuvant-adjuvant therapy.
  • the anti-MUC1 antibody is for the treatment of a patient as palliative therapy.
  • the cancer therapy with the anti-MUC1 antibody preferably results in inhibition of tumor growth and in particular reduction of tumor size. Furthermore, the occurrence of further metastases is prevented and/or their number is reduced by the treatment.
  • the treatment preferably results in an increase in progression-free survival; and/or an increase in lifespan and thus the overall survival.
  • the present invention further provides methods of therapy, diagnosis, prognosis, detecting and/or monitoring of a disease using the antibody according to the invention.
  • the embodiments and examples of the use of the antibody in medicine also apply likewise to the medical methods.
  • a method for treating a disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the antibody according to the present invention is provided.
  • the invention provides a method for treating cancer in a subject in need thereof comprising, administering to the subject with cancer a therapeutically effective amount of the antibody according to the invention.
  • the cancer is characterized by expressing TA-MUC1.
  • the cancer may be selected from the group consisting of ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemia, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
  • the invention provides a method for diagnosis, detecting or monitoring cancer, comprising the step of contacting a test sample with an antibody according to the invention.
  • the invention provides a method of increasing the MUC1 binding affinity of an antibody comprising
  • the method comprising the step of substituting the amino acid residue at position 8 of CDR-H2 with any amino acid residue except asparagine, resulting in CDR-H2 having the amino acid sequence of SEQ ID NO: 2.
  • the antibody which MUC1 binding affinity is to be increased in particular is an antibody capable of binding to MUC1 as described herein, except that it comprises an asparagine at position 8 of the CDR-H2 sequence.
  • the heavy chain variable region of the antibody which MUC1 binding affinity is to be increased comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 11.
  • the heavy chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 11.
  • the heavy chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 1, 8 and 3. Hence, any sequence deviations to SEQ ID NO: 11 are located in the framework regions, but not in the CDRs.
  • the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 11.
  • the light chain variable region of the antibody which MUC1 binding affinity is to be increased comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12.
  • the light chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 12.
  • the light chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 4, 5 and 6. Hence, any sequence deviations to SEQ ID NO: 12 are located in the framework regions, but not in the CDRs.
  • the light chain variable region comprises the amino acid sequence of SEQ ID NO: 12.
  • the heavy chain variable region of the antibody which MUC1 binding affinity is to be increased has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 11, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 8 and 3, and the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the heavy chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 11, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 8 and 3 and the light chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • the antibody which MUC1 binding affinity is to be increased is an anti-MUC1 antibody as disclosed in WO 2004/065423 A2 or WO 2011/012309 A1.
  • the antibody which MUC1 binding affinity is to be increased is gatipotuzumab or PankoMab.
  • the antibody which MUC1 binding affinity is increased in particular is an antibody capable of binding to MUC1 as described herein.
  • MUC1 binding is as described herein.
  • Increasing the MUC1 or TA-MUC1 binding affinity in particular refers to an increase of at least 10%, at least 20%, at least 33% or at least 50%.
  • MUC1 binding affinity is increased by at least 50%.
  • the MUC1 binding affinity may be determined as described in the examples, especially using surface plasmon resonance analysis or switchSENSE® Technology (DRX2 Biosensor, manufactured by Dynamic Biosensors GmbH), as described, e.g., in example 4a and b.
  • the step of substituting the amino acid residue at position 8 of CDR-H2 is achieved by introducing a mutation into the nucleic acid coding for the antibody, wherein the mutation is introduced in the codon coding for said amino acid residue.
  • Introducing the mutation can be done by any method. Several suitable methods are known in the art and the skilled person is capable of performing the necessary tasks to introduce the mutation.
  • the antibody with increased MUC1 binding affinity can then be obtained by expressing the mutated nucleic acid, for example in a host cell. Nucleic acids, host cells and methods for producing the antibody are described herein and can be used for the method for increasing the MUC1 binding affinity.
  • the method of increasing the MUC1 binding affinity of an antibody comprises the steps of
  • the present invention further provides a method of producing an antibody with increased MUC1 binding affinity, comprising
  • the method of producing an antibody with increased MUC1 binding affinity further comprises a step (d) of processing the antibody with increased MUC1 binding affinity.
  • processing the antibody with increased MUC1 binding affinity may include isolating the antibody from the cell culture.
  • Isolation of the antibody in particular refers to the separation of the antibody from the remaining components of the cell culture. Separation of the antibody from the cell culture medium may be performed, for example, by chromatographic methods. Suitable methods and means for isolating antibodies are known in the art and can be readily applied by the skilled person.
  • the obtained antibody may optionally be subject to further processing steps such as e.g. modification steps such as chemical or enzymatic coupling of a further agent to the antibody, and/or formulation steps in order to produce the antibody in the desired quality and composition.
  • further processing steps and methods are generally known in the art.
  • step (d) additionally comprises the step of providing a pharmaceutical formulation comprising the antibody.
  • Providing a pharmaceutical formulation comprising the antibody or formulating the antibody as a pharmaceutical composition in particular comprises exchanging the buffer solution or buffer solution components of the composition comprising the antibody.
  • this step may include lyophilization of the antibody.
  • the antibody is transferred into a composition only comprising pharmaceutically acceptable ingredients.
  • Embodiment 1 An antibody capable of binding to MUC1, which comprises
  • Embodiment 2 The antibody according to Embodiment 1, wherein the amino acid at position 8 of the CDR-H2 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine, especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine, in particular glutamine.
  • Embodiment 3 The antibody according to Embodiment 1, wherein the amino acid at position 8 of the CDR-H2 is glutamine, histidine, arginine, tryptophan, or lysine.
  • Embodiment 4 The antibody according to Embodiments 1 to 3, wherein the CDR-H2 has the amino acid sequence of SEQ ID NO: 7.
  • Embodiment 6 An antibody capable of binding to MUC1, which comprises
  • Embodiment 7 An antibody capable of binding to MUC1, which comprises
  • Embodiment 8 The antibody according to Embodiment 6 or 7, wherein the amino acid at position 8 of the CDR-H2 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine, especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine, in particular glutamine.
  • Embodiment 9 The antibody according to Embodiment 7 or 8, wherein the amino acid at position 8 of the CDR-H2 is glutamine, histidine, arginine, tryptophan, or lysine.
  • Embodiment 10 An antibody capable of binding to MUC1, which comprises
  • Embodiment 11 An antibody capable of binding to MUC1, which comprises
  • Embodiment 12 An antibody capable of binding to MUC1, which comprises
  • Embodiment 13 The antibody according to Embodiment 12, wherein the amino acid at position 57 of SEQ ID NO: 9 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine, especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine, in particular glutamine.
  • Embodiment 14 The antibody according to Embodiment 12, wherein the amino acid at position 57 of SEQ ID NO: 9 is glutamine, histidine, arginine, tryptophan, or lysine.
  • Embodiment 15 An antibody capable of binding to MUC1, which comprises
  • Embodiment 16 An antibody capable of binding to MUC1, which comprises
  • Embodiment 17 An antibody capable of binding to MUC1, which comprises
  • Embodiment 18 An antibody capable of binding to MUC1, which comprises
  • Embodiment 19 An antibody capable of binding to MUC1, which comprises
  • Embodiment 20 An antibody capable of binding to MUC1, which comprises
  • Embodiment 21 An antibody capable of binding to MUC1, which comprises
  • Embodiment 22 The antibody according to any one of Embodiments 1 to 21, wherein the antibody comprises at least one heavy chain, comprising the heavy chain variable region, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain.
  • Embodiment 23 The antibody according to any one of Embodiments 1 to 21, wherein the antibody comprises two heavy chains, each comprising the heavy chain variable region, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain.
  • Embodiment 24 The antibody according to Embodiment 22 or 23, wherein the antibody is an IgG-type antibody, in particular an IgG1, IgG2 or IgG4-type antibody.
  • Embodiment 25 The antibody according to any one of Embodiments 1 to 24, wherein the antibody comprises at least one light chain, comprising the light chain variable region and a CL domain.
  • Embodiment 26 The antibody according to any one of Embodiments 1 to 24, wherein the antibody comprises two light chains, each comprising the light chain variable region and a CL domain.
  • Embodiment 274 The antibody according to Embodiment 25 or 26, wherein the light chain is a ⁇ -type light chain.
  • Embodiment 28 The antibody according to any one of Embodiments 1 to 27, wherein the antibody does not comprise an N-glycosylation site in the CH2 domain.
  • Embodiment 29 The antibody according to any one of Embodiments 1 to 27, wherein the antibody comprises an N-glycosylation site in the CH2 domain of the antibody heavy chains.
  • Embodiment 30 The antibody according to Embodiment 29, wherein the antibody has a glycosylation pattern having one or more of the following characteristics
  • Embodiment 31 The antibody according to Embodiment 29, wherein the antibody has a glycosylation pattern having one or more of the following characteristics
  • Embodiment 32 The antibody according to any one of Embodiments 1 to 31, comprising a further agent conjugated thereto.
  • Embodiment 33 The antibody according to Embodiment 32, wherein the further agent is a chemotherapeutic agent which is coupled to the antibody.
  • Embodiment 34 The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of a maytansinoid, a DNA damaging agent, a DNA alkylating agent and a DNA minor groove binder.
  • the chemotherapeutic agent is selected from the group consisting of a maytansinoid, a DNA damaging agent, a DNA alkylating agent and a DNA minor groove binder.
  • Embodiment 35 The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of maytansinol, N 2′ -deacetyl-N 2′ -(3-mercapto-1-oxopropyl)-maytansine (DM1), N 2′ -deacetyl-N 2′ -(4-mercapto-1-oxopentyl)-maytansine (DM3), and N 2′ -deacetyl-N 2′ -(4-methyl-4-mercapto-1-oxopentyl)-maytansine (DM4).
  • DM1 maytansinol
  • DM3 maytansinol
  • DM3 N 2′ -deacetyl-N 2′ -(4-mercapto-1-oxopentyl)-maytansine
  • DM4 N 2′ -deacetyl-N 2′ -(4-methyl-4-mer
  • Embodiment 36 The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of monomethyl auristatin F (MMAF), monomethyl auristatin E (MMAE) and auristatin T.
  • MMAF monomethyl auristatin F
  • MMAE monomethyl auristatin E
  • T auristatin T
  • Embodiment 37 The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of pyrrolobenzodiazepine (PBD), pyrrolobenzodiazepine dimer (PBD dimer), duocarmycin, duocarmycin-hydroxybenzamide-azaindole (DUBA), seco-duocarmycin-hydroxybenzamide-azaindole (seco-DUBA) and doxorubicin.
  • PBD pyrrolobenzodiazepine
  • PBD dimer pyrrolobenzodiazepine dimer
  • duocarmycin duocarmycin-hydroxybenzamide-azaindole
  • DUBA duocarmycin-hydroxybenzamide-azaindole
  • seco-DUBA seco-duocarmycin-hydroxybenzamide-azaindole
  • doxorubicin doxorubicin.
  • Embodiment 38 The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of indolinobenzodiazepine and oxazolidinobenzodiazepine.
  • Embodiment 39 The antibody according to Embodiment 33, wherein the chemotherapeutic agent is calicheamicin.
  • Embodiment 40 The antibody according to Embodiment 32, wherein the further agent is a polypeptide or protein which is fused to a polypeptide chain of the antibody.
  • Embodiment 41 The antibody according to Embodiment 40, wherein the antibody comprises two antibody heavy chains and two antibody light chains and a further agent being a polypeptide or protein is fused to each of the C termini of said antibody heavy chains or to each of the C termini of said antibody light chains.
  • Embodiment 42 The antibody according to Embodiment 40 or 41, wherein the further agent is selected from the group consisting of cytokines, chemokines, other antibodies, antigen binding fragments, enzymes and binding domains.
  • Embodiment 43 The antibody according to Embodiment 41, wherein the further agent is a scFv fragment specifically binding to CD3, and one of said further agent is fused to the C terminus of each antibody heavy chain.
  • Embodiment 44 The antibody according to Embodiment 41, wherein the further agent is a scFv fragment specifically binding to PDL1, and one of said further agent is fused to the C terminus of each antibody light chain.
  • Embodiment 45 A nucleic acid encoding the antibody according to any one of Embodiments 1 to 44.
  • Embodiment 46 An expression cassette or vector comprising the nucleic acid according to Embodiment 75 and a promoter operatively connected with said nucleic acid.
  • Embodiment 47 A host cell comprising the nucleic acid according to Embodiment 45 or the expression cassette or vector according to Embodiment 46.
  • Embodiment 48 A pharmaceutical composition comprising the antibody or conjugate according to any one of Embodiments 1 to 44 and one or more further components selected from the group consisting of solvents, diluents, and excipients.
  • Embodiment 49 The antibody according to any one of Embodiments 1 to 44 or the pharmaceutical composition according to Embodiment 48 for use in medicine.
  • Embodiment 50 The antibody according to any one of Embodiments 1 to 44 or the pharmaceutical composition according to Embodiment 48 for use in the treatment, prognosis, diagnosis, detecting and/or monitoring of diseases associated with abnormal cell growth such as cancer; infections such as bacterial, viral, fungal or parasitic infections; inflammatory diseases such as autoimmune diseases and inflammatory bowel diseases; and diseases associated with a reduce immune activity such as immunodeficiencies.
  • diseases associated with abnormal cell growth such as cancer
  • infections such as bacterial, viral, fungal or parasitic infections
  • inflammatory diseases such as autoimmune diseases and inflammatory bowel diseases
  • diseases associated with a reduce immune activity such as immunodeficiencies.
  • Embodiment 51 The antibody or pharmaceutical composition according to Embodiment 50 for use in the treatment of cancer, in particular the cancer expressing TA-MUC1, wherein the cancer is selected from the group consisting of cancer of the ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemia, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
  • the cancer is selected from the group consisting of cancer of the ovarian cancer, breast cancer, pancreatic
  • Embodiment 52 The antibody or pharmaceutical composition according to Embodiment 50 for use in the treatment of infections, wherein the infection is selected from the group consisting of bacterial infections, viral infections, fungal infections and parasitic infections.
  • Embodiment 53 The antibody or pharmaceutical composition according to Embodiment 50 for use in the treatment of autoimmune diseases, wherein the autoimmune disease is selected from the group consisting of celiac disease, diabetes mellitus type 1, Graves disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis and systemic lupus erythematosus.
  • the autoimmune disease is selected from the group consisting of celiac disease, diabetes mellitus type 1, Graves disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis and systemic lupus erythematosus.
  • Embodiment 54 A method of increasing the MUC1 binding affinity of an antibody comprising
  • the method comprising the step of substituting the amino acid residue at position 8 of CDR-H2 with any amino acid residue except asparagine, resulting in CDR-H2 having the amino acid sequence of SEQ ID NO: 2.
  • Embodiment 55 A method of increasing the MUC1 binding affinity of an antibody comprising
  • the method comprising the step of substituting the amino acid residue at position 8 of CDR-H2 with any amino acid residue except asparagine, resulting in CDR-H2 having the amino acid sequence of SEQ ID NO: 2.
  • Embodiment 56 The method according to embodiment 54 or 55, wherein substituting the amino acid residue at position 8 of CDR-H2 is achieved by introducing a mutation into the nucleic acid coding for the antibody, wherein the mutation is introduced in the codon coding for said amino acid residue.
  • Embodiment 57 The method according to any one of Embodiments 54 to 56, comprising the steps of
  • Embodiment 58 A method of producing an antibody with increased MUC1 binding affinity, comprising
  • Embodiment 59 A method of producing an antibody with increased MUC1 binding affinity, comprising
  • Embodiment 60 The method according to any one of Embodiments 54 to 59, wherein the antibody comprises two heavy chains, each comprising the heavy chain variable region, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain.
  • Embodiment 61 The method according to Embodiment 60, wherein the antibody is an IgG-type antibody, in particular an IgG1, IgG2 or IgG4-type antibody.
  • Embodiment 62 The method according to any one of Embodiments 54 to 60, wherein the antibody comprises two light chains, each comprising the light chain variable region and a CL domain.
  • Embodiment 63 The method according to Embodiment 62, wherein the light chain is a ⁇ -type light chain.
  • Embodiment 64 The method according to any one of Embodiments 54 to 63, wherein the antibody with increased MUC1 binding affinity is an antibody as defined in any one of Embodiments 1 to 44.
  • Embodiment 65 A method for treating cancer in a subject in need thereof comprising, administering to the subject with cancer, in particular cancer expressing TA-MUC1, a therapeutically effective amount of the antibody according to any one of Embodiments 1 to 44 or the composition according to Embodiment 48.
  • Embodiment 66 The method for treating cancer according to Embodiment 65, wherein the cancer is selected from the group consisting of ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemia, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
  • the cancer is selected from the group consisting of ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial
  • FIG. 1 shows ELISA binding curves of the anti-MUC1 antibodies to different MUC1 peptides.
  • A shows antigen binding of PankoMab N54Q (PM-N54Q) lacking Fab glycosylation and PankoMab comprising Fab glycosylation (PM) to the MUC1 peptide comprising the epitope sequence PDTR.
  • the threonine of the MUC1 peptide is glycosylated with Tn, sTn, TF or sTF.
  • B shows binding of PankoMab and PankoMab N54Q to the MUC1 peptide comprising the epitope sequence variant PESR.
  • the serine of the MUC1 peptide is glycosylated with Tn.
  • C shows binding of PankoMab N54Q to the MUC1 peptide comprising the epitope sequence PDTR. The threonine of the MUC1 peptide is glycosylated with Tn or not glycosylated.
  • D shows binding of several N54X variants to Tn-PDTR MUC1 peptide compared to PankoMab comprising Fab glycosylation diluted from cell culture supernatant of transiently transfected cells.
  • (E) shows binding curves of three purified N54X variants without Fab glycosylation in comparison to PankoMab with Fab glycosylation on Tn-PDTR, TF-PDTR and non-glycosylated PDTR MUC1 peptide.
  • (F) shows binding of two framework variants of PM-N54Q to Tn-PDTR MUC1 peptide compared to PankoMab with Fab glycosylation.
  • For framework variant mf-a nine amino acids are mutated in the VH and three in the VL framework, for mf-b also nine amino acids are mutated in the VH and four in the VL framework.
  • FIG. 2 shows surface plasmon resonance (Biacore) binding of the anti-MUC1 antibodies PM and PM-N54Q to a Tn-glycosylated PDTR-MUC1 peptide.
  • the maximal binding signal of different concentrations of PankoMab N54Q and PankoMab are plotted against the antibody concentration.
  • FIG. 3 shows results of Fluorescence Proximity Sensing on DRX instrument. Association and dissociation curves are shown.
  • A PM with Fab glycosylation compared to
  • B PM-N54Q without Fab glycosylation.
  • FIG. 4 shows an SDS acrylamide gel of an electrophoretic separation of PankoMab N54Q and PankoMab under non-reducing (left) and reducing (right) conditions.
  • Lane 1 PankoMab N54Q after capture step
  • lane 2 PankoMab N54Q after polishing step
  • lane 3 PankoMab after capture step
  • lane 4 PankoMab after polishing step
  • lane 5 molecular weight marker.
  • FIG. 5 shows the Coomassie blue stained gel of an isoelectric focusing assay with PankoMab N54Q lacking Fab glycosylation and PankoMab being Fab-glycosylated.
  • Lane 1 PankoMab with Fab glycosylation
  • lane 2 PankoMab N54Q without Fab glycosylation.
  • FIG. 6 shows anti-MUC1 antibody binding to Fc ⁇ receptor IIIa. Increasing concentrations of the antibody PankoMab N54Q or PankoMab displace rabbit-anti-mouse coupled acceptor beads from Fc ⁇ RIIIa loaded donor beads, thereby reducing the detected chemiluminescence. In FIG. 6A low-fucosylated antibodies and in FIG. 6B high-fucosylated antibodies were applied into the assay.
  • FIG. 7 shows binding of the anti-MUC1 antibodies PM-N54Q, PM-N54D and PM with Fab glycosylation to the tumor cell lines (A) CaOV-3 and (B) HSC-4 as analyzed by flow cytometry.
  • FIG. 8 shows the amino acid sequence of the heavy chain of the humanized antibody PankoMab N54Q (SEQ ID No: 15, wherein the amino acid at position 57 is Gln, namely SEQ ID No: 22).
  • FIG. 9 shows the amino acid sequence of the light chain of the humanized antibody PankoMab N54Q (SEQ ID No: 16).
  • FIG. 10 shows the amino acid sequence of the heavy chain of the humanized antibody PankoMab (SEQ ID No: 19).
  • FIG. 11 shows the amino acid sequence of the heavy chain of chimeric antibody PankoMab N54Q (SEQ ID No: 20, wherein the amino acid at position 76 is Gln, namely SEQ ID No: 23).
  • FIG. 12 shows the amino acid sequence of the light chain of chimeric antibody PankoMab N54Q (SEQ ID No: 21).
  • the nucleic acid sequence of the heavy chain of humanized PankoMab antibody was modified by mutating the codon for Asn54 according to the Kabat/EU numbering system (amino acid position 57 in SEQ ID NO: 11) into the codon for any amino acid except Asn, especially for Gln.
  • Vectors comprising the coding sequences of the ⁇ 1-type heavy chain and the ⁇ -type light chain of the mutated antibodies were transfected into the human myeloid leukemia derived cell line NM-H9D8 (DSM ACC2806).
  • the different ⁇ MUC1-antibodies comprising the N54X mutation (PankoMab N54X/PM-N54X whereby X is any amino acid except N/Asn) or amino acid mutations in the framework sequences of the VH and VL were expressed in the obtained clones, producing the constructs with a human glycosylation pattern.
  • the concentration of the ⁇ MUC1-antibodies in the supernatant was determined by Octet measurement using Protein A coated pins or were quantified by UV280 absorbance after purification by protein A chromatography.
  • the binding characteristics of the different ⁇ MUC1-antibodies were determined by Antigen-ELISA (see example 2), and selected purified antibodies were also analyzed by Scatchard analysis (see example 3), by Biacore (see example 4a), by DRX 2 switchSENSE® technology (see example 4b), or by flow cytometry (example 7).
  • PM-N54Q and non-mutated PankoMab with Fab-glycosylation were also expressed in the human myeloid leukemia derived cell line NM-H9D8-E6Q12 (DSM ACC2856) expressing antibody with reduced fucose. Together with the same antibodies expressed in NM-H9D8, these antibodies were purified and analyzed in example 6 for their binding behavior to Fc gamma receptor Ill A.
  • PM-N54Q encoding sequences (nucleotide sequence of heavy chain of PM-N54Q represented by SEQ ID NO: 17 and nucleotide sequence of light chain of PM-N54Q represented by SEQ ID NO: 18) which was synthesized by GeneArtTM of ThermoFisher scientific were cloned into expression vectors and resulting plasmids were electro-transfected into CHO cells. Pooled cells grown under selection pressure were applied to manufacture PM-N54Q mutant antibody with general procedures.
  • PankoMab N54X wherein the N-glycosylation site in the Fab part is knocked out, was compared to PankoMab having an N-glycosylation site in its Fab part.
  • Binding characteristics of the Fab-deglycosylated version of the MUC1-specific antibody PankoMab (PM-N54Q) compared to the (glycosylated) PankoMab-GEX® were analyzed using differently glycosylated and the non-glycosylated MUC1-derived tandem repeat peptides in ELISA studies.
  • both antibodies show the same gradation by means of binding to glycosylated PDTR peptides (APPAHGVTSAPD-T(X)-RPAPGSTAPPAHGVTSA) with different glycosylations at T: Strongest binding was observed to the PDTR peptide carrying a Gal ⁇ 1-3GalNAc alpha (TF) followed by sialylated TF GalNAc alpha (Tn) O-glycosylation. Binding to sialylated GalNAc alpha (sTn)O-glycosylation was significantly lower. As PankoMab-GEX®, PM-N54Q showed only little binding activity to non-glycosylated MUC1 PDTR peptide indicating adequate tumor specificity ( FIG. 1C ).
  • PM-N54Q binds about seven-fold better to the same MUC1 peptide when glycosylated with sialylated GalNAc alpha (sTn).
  • the framework variant mf-a carries nine amino acid mutations in the VH and three in the VL framework; the variant mf-b carries also nine amino acid mutations in the VH and four in the VL framework. Both mutated variants show similar binding compared to the PM-N54Q antibody.
  • Binding of the Fab-deglycosylated version of the MUC1-specific antibody PankoMab (PM-N54Q) on TA-MUC-1 positive human tumor cell lines was evaluated using radiolabeled antibodies by saturated binding analysis on the human mamma carcinoma cell lines ZR-75-1 and MCF-7 in comparison to Fab-glycosylated PankoMab-GEX®.
  • the antibodies were chelated with a 12-fold molar excess of p-SCN-Benzyl-DTPA in 50 mM sodium carbonate, 150 mM NaCl, pH 8.7, for 2 h at 37° C., followed by over-night incubation at 2-8° C. Free chelator was removed over desalting columns and dead-end filtration (50 kDa cut-off, 6 ⁇ buffer exchange to PBS).
  • the chelated antibodies were radiolabeled with carrier-free 111 In (2 ⁇ Ci/ ⁇ g antibody) for 1 h at 37° C. in 6 mM phosphate, 1.6 mM KCl, 80 mM NaCl, 0.2 M Na-acetate, 0.1 M HCl.
  • the preparations were neutralized by addition of 8-9fold volume of 10 ⁇ concentrated PBS. About 1/50 volume of fetal bovine serum were added to the neutralized labelled antibody preparation. Per cell binding approach 1*10 6 cells were used.
  • concentrations of labelled antibodies were added to the pelleted cells (30-1000 ng/200 ⁇ L in 1% BSA/PBS). The resuspended cell-antibody mixtures were measured in a gamma-counter and incubated 1 h at 4° C. Cells with bound antibodies were separated by centrifugation and washed with 1% BSA/PBS for another hour at 4° C. The cell pellet was then measured for bound 111 In-labelled antibody in a gamma counter.
  • PankoMab (PM-N54Q) on TA-MUC-1 derived glycosylated peptide was evaluated by surface plasmon resonance analysis (Biacore).
  • a streptavidin sensor chip was coated with biotinylated TA-MUC1 peptide (Tn glycosylated or not glycosylated).
  • PankoMab and PM-N54Q were diluted sequentially 1:3 from 3,600 to 4.9 nM in HPS-EP. The dilutions were injected at 50 ⁇ L/min.
  • FIG. 2 shows the obtained binding curves with PM-N54Q compared to PankoMab-GEX®. Affinities (K) of 388 nM and 652 nM were calculated for PM-N54Q and PankoMab-GEX®, respectively. Therefore, in this experimental setting a nearly two-fold increase in affinity was detectable.
  • Example 4b Fluorescence Proximity Sensing (by DRX 2 , Dynamic Biosensors)
  • a new method to determine binding constants and affinity is the fluorescence proximity sensing using single stranded DNA (96mer) spotted on a chip and complementary DNA coupled to a ligand.
  • streptavidin was used as a ligand to capture biotinylated TA-MUC1 peptides.
  • Binding of PankoMab to the peptides resulted in a fluorescence change.
  • On- and off-rates can be calculated during association and dissociation. Due to a higher sensitivity faster interactions can be monitored compared to surface plasmon resonance. This results in binding kinetics different from SPR but more comparable to the “gold standard” method KinExA, measured in a liquid system.
  • PankoMab and PM-N54Q were diluted from 300 nM in 1:9 steps to 3.67 nM in PE140 buffer and applied to the chip-bound peptides. Binding curves were evaluated by mono-exponential global fit (instrument software). Binding curves of PM and PM-N54Q are exemplarily shown in FIGS. 3A and B. Calculated affinities of PankoMab variants are shown in Table 2:
  • Non-reducing and reducing SDS-PAGE is used to analyze purity and identity of an antibody.
  • the band pattern in non-reducing gels shows the major band at about 160 kDa and methodical artefacts of heavy and light chains and combinations thereof (25, 50-55, 75, 110, 135 kDa).
  • Reducing gels show distinct light and heavy chain bands at 25 and 50-55 kDa. Due to lack of the Fab glycosylation PM-N54Q has a smaller heavy chain, as expected (see FIG. 4 , right).
  • Fc ⁇ R binding assays for Fc ⁇ RIIIa are based on the AlphaScreen® technology of PerkinElmer.
  • the AlphaScreen® platform relies on simple bead-based technology of PerkinElmer and is a more efficient alternative to traditional ELISA since no washing steps are necessary.
  • Fc ⁇ RIIIa His-tagged Fc ⁇ RIIIa (Glycotope GmbH) is captured by Ni-chelate donor beads.
  • Anti-MUC1 antibodies and rabbit-anti-mouse coupled acceptor beads compete for binding to Fc ⁇ R.
  • donor and acceptor beads come into close proximity which leads, upon laser excitation at 680 nm, to light emission.
  • a maximum signal is achieved (signal max ) without a competitor.
  • the signal max is reduced in a concentration-dependent manner.
  • Chemiluminescence was quantified by measurement at 520-620 nm (AlphaScreen® method) using an EnSpire 2300 multilabel reader (PerkinElmer). All results were expressed as the mean ⁇ standard deviation of duplicate samples. The data were evaluated and calculated using non-linear curve fitting (sigmoidal dose-response variable slope) with GraphPad Prism 5 software. As a result, a concentration dependent sigmoidal curve was obtained, which is defined by top-plateau, bottom-plateau, slope and EC 50 .
  • the Fc ⁇ RIIIa binding affinity was comparable for PankoMab N54Q and PankoMab whereby in Figure A low-fucosylated antibodies and in Figure B high-fucosylated antibodies were applied into the assay. Hence, removal of the Fab glycosylation did not affect receptor interaction of the antibody.
  • N54Q and N54D were transiently expressed and purified by protein A chromatography. Binding of the two variants to cell surface TA-MUC1 was compared to PM with Fab glycosylation using two different carcinoma cell lines.
  • the tongue squamous cell carcinoma line HSC-4 expresses TA-MUC1 to a medium degree and the ovarian carcinoma cell line CaOV-3 to a high degree.
  • Tumor cells were incubated with antibodies in serial dilutions and bound antibodies were detected using a Phycoerythrin-conjugated goat anti-human IgG (heavy and light chain) antibody. A human IgG control was included to control for background staining. Binding was analyzed by flow cytometry.
  • the analyzed constructs PM, PM-N54Q and PM-N54D show strong and specific binding to the TA-MUC1 expressing HSC-4 and CaOV-3 cells compared to a human IgG1 control ( FIG. 7 ).
  • the binding of PM-N54D to the TA-MUC1 high CaOV-3 cells was comparable to PM with Fab glycosylation while PM-N54Q showed a slightly better binding ( FIG. 7A ).
  • HSC-4 carcinoma cells that express TA-MUC1 at an intermediate level the variant PM-N54Q was clearly superior in binding to cellular TA-MUC1 compared to PM while PM-N54D showed an inferior binding compared to PM with Fab glycosylation ( FIG. 7B ).
  • the cell lines DSM ACC 2806, DSM ACC 2807 and DSM ACC 2856 were deposited at the DSMZ—Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH, Inhoffenstralße 7B, 38124 Braunschweig (DE) by Glycotope GmbH, Robert-Rössle-Str. 10, 13125 Berlin (DE) on the dates indicated in the following table.

Abstract

The present disclosure pertains to novel antibodies directed against the cancer antigen MUC1. In particular, an antibody with improved antigen binding was obtained by deleting a glycosylation site in the CDR-H2 of a known anti-MUC1 antibody.

Description

    FIELD OF THE INVENTION
  • The present invention pertains to the field of antibodies. A mutated anti-MUC1 antibody with increased antigen binding affinity is provided. In particular, a mutated version of the humanized antibody PankoMab is provided wherein asparagine 57 of the heavy chain variable region is substituted by another amino acid. Thereby, the glycosylation site in the CDR2 region is deleted and the antigen binding affinity is increased. In specific embodiments, the present invention is directed to the therapeutic and diagnostic use of this antibody and to methods of producing such antibodies.
  • BACKGROUND OF THE INVENTION
  • Antibodies against tumor-associated antigens are widely used therapeutics against cancers. Today, many anti-cancer antibodies are approved for human therapy. Some of these antibodies act by blocking certain signaling pathways which are critical for survival or proliferation of specific cancer cells. Other anti-cancer antibodies activate the patient's immune response against the targeted cancer cells, for example by initiating antibody-dependent cellular cytotoxicity (ADCC) via natural killer cells. This mechanism is induced by binding of the antibody's Fc part to Fc receptors on the immune cells.
  • An interesting and important group of antibodies are those directed against mucin proteins. Mucins are a family of high molecular weight, heavily glycosylated proteins produced by many epithelial tissues in vertebrates. They can be subdivided into mucin proteins which are membrane-bound due to the presence of a hydrophobic membrane-spanning domain that favors retention in the plasma membrane, and mucins which are secreted onto mucosal surfaces or secreted to become a component of saliva. The human mucin protein family consists of many family members, including membrane bound MUC1.
  • Increased mucin production occurs in many adenocarcinomas, including cancer of the pancreas, lung, breast, ovary, colon, etc. Mucins are also overexpressed in lung diseases such as asthma, bronchitis, chronic obstructive pulmonary disease or cystic fibrosis. Two membrane mucins, MUC1 and MUC4 have been extensively studied in relation to their pathological implication in the disease process. Moreover, mucins are also being investigated for their potential as diagnostic markers. Several antibodies directed against mucin proteins (Clin. Cancer Res., 2011 Nov. 1;17(21):6822-30, PLoS One, 2011 Jan. 14;6(1):e15921), in particular MUC1, are known in the art. However, their therapeutic efficacy could still be improved.
  • In view of this, there is a need in the art to provide therapeutic anti-MUC1 antibodies with improved properties.
  • SUMMARY OF THE INVENTION
  • The present inventors have found that deleting the glycosylation site in the heavy chain variable region of the anti-MUC1 antibody PankoMab did not abolish antigen binding, but rather unexpectedly increased the antigen affinity of the antibody. This was in particular surprising as the glycosylation site is located in the second complementarity-determining region of the heavy chain variable region (CDR-H2). The CDRs are those regions of an antibody which are directly involved in antigen binding and provide the contact to the epitope. Therefore, generally modifying the amino acids of a CDR is expected to be detrimental to the antigen binding affinity. The humanized PankoMab antibody additionally comprises a glycosylation site in CDR-H2, which carries a large carbohydrate structure. This carbohydrate structure is present directly at the binding interface to the antigen and hence, was considered to be involved in antigen binding.
  • However, as demonstrated in the examples, the PankoMab variant (N54Q) wherein the glycosylation site is deleted by substituting the amino acid carrying the carbohydrate structure exhibits an increased antigen binding affinity.
  • Therefore, in a first aspect, the present invention is directed to an antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • In a second aspect, the present invention provides a nucleic acid encoding the antibody according to the invention. Furthermore, in a third aspect an expression cassette or vector comprising the nucleic acid according to the invention and a promoter operatively connected with said nucleic acid and, in a fourth aspect, a host cell comprising the nucleic acid or the expression cassette or vector according to the invention are provided.
  • In a fifth aspect, the present invention provides a conjugate comprising the antibody according to the invention conjugated to a further agent.
  • In a sixth aspect, the present invention is directed to a composition comprising the antibody according to the invention, the nucleic acid according to the invention, the expression cassette or vector according to the invention, the host cell according to the invention, or the conjugate according to the invention.
  • According to a seventh aspect, the invention provides the antibody, the nucleic acid, the expression cassette or vector, the host cell, the composition or the conjugate according to the invention for use in medicine, in particular in the treatment, prevention or diagnosis of cancer.
  • In an eighth aspect, the invention provides a method of increasing the MUC1 binding affinity of an antibody comprising
      • (i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6,
  • the method comprising the step of substituting the amino acid residue at position 8 of CDR-H2 with any amino acid residue except asparagine, resulting in CDR-H2 having the amino acid sequence of SEQ ID NO: 2.
  • In a ninth aspect, the invention provides a method of producing an antibody with increased MUC1 binding affinity, comprising
      • (a) providing a nucleic acid coding for an antibody which comprises
        • (i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
        • (ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6;
      • (b) introducing a mutation into said nucleic acid to produce a mutated nucleic acid, wherein the mutation is introduced in the codon coding for the amino acid residue at position 8 of CDR-H2 so that said codon codes for any amino acid residue except asparagine; and
      • (c) producing the antibody with increased MUC1 binding affinity by expressing the mutated nucleic acid in a host cell.
  • In a tenth aspect, the present invention provides a method for treating cancer in a subject in need thereof comprising, administering to the subject with cancer a therapeutically effective amount of the antibody according to the invention, the nucleic acid according to the invention, the expression cassette or vector according to the invention, or the host cell according to the invention.
  • In a eleventh aspect, the present invention provides kits or devices comprising the antibody according to the invention, and associated methods that are useful in the diagnosis, detecting or monitoring of MUC1 associated disorders such as cancer.
  • Other objects, features, advantages and aspects of the present invention will become apparent to those skilled in the art from the following description and appended claims.
  • It should be understood, however, that the following description, appended claims, and specific examples, which indicate preferred embodiments of the application, are given by way of illustration only. Various changes and modifications within the spirit and scope of the disclosed invention will become readily apparent to those skilled in the art from reading the following.
  • Definitions
  • As used herein, the following expressions are generally intended to preferably have the meanings as set forth below, except to the extent that the context in which they are used indicates otherwise.
  • The expression “comprise”, as used herein, besides its literal meaning also includes and specifically refers to the expressions “consist essentially of” and “consist of”. Thus, the expression “comprise” refers to embodiments wherein the subject-matter which “comprises” specifically listed elements does not comprise further elements as well as embodiments wherein the subject-matter which “comprises” specifically listed elements may and/or indeed does encompass further elements. Likewise, the expression “have” is to be understood as the expression “comprise”, also including and specifically referring to the expressions “consist essentially of” and “consist of”. The term “consist essentially of”, where possible, in particular refers to embodiments wherein the subject-matter comprises 20% or less, in particular 15% or less, 10% or less or especially 5% or less further elements in addition to the specifically listed elements of which the subject-matter consists essentially of.
  • The term “antibody” in particular refers to a protein comprising at least two heavy chains and two light chains connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (VH) and a heavy chain constant region (CH). Each light chain is comprised of a light chain variable region (VL) and a light chain constant region (CL). The heavy chain-constant region comprises three or—in the case of antibodies of the IgM- or IgE-type—four heavy chain-constant domains (CH1, CH2, CH3 and CH4) wherein the first constant domain CH1 is adjacent to the variable region and may be connected to the second constant domain CH2 by a hinge region. The light chain-constant region consists only of one constant domain. The variable regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR), wherein each variable region comprises three CDRs and four FRs. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The heavy chain constant regions may be of any type such as γ-, δ-, α-, μ- or ε-type heavy chains. Preferably, the heavy chain of the antibody is a γ-chain. Furthermore, the light chain constant region may also be of any type such as κ- or λ-type light chains. Preferably, the light chain of the antibody is a κ-chain. The terms “γ- (δ-, α-, μ- or ε-) type heavy chain” and “κ- (λ-) type light chain” refer to antibody heavy chains or antibody light chains, respectively, which have constant region amino acid sequences derived from naturally occurring heavy or light chain constant region amino acid sequences, especially human heavy or light chain constant region amino acid sequences. In particular, the amino acid sequence of the constant domains of a γ-type (especially γ1-type) heavy chain is at least 95%, especially at least 98%, identical to the amino acid sequence of the constant domains of a human γ (especially the human γ1) antibody heavy chain. Furthermore, the amino acid sequence of the constant domain of a κ-type light chain is in particular at least 95%, especially at least 98%, identical to the amino acid sequence of the constant domain of the human K antibody light chain. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system. The antibody can be e.g. a humanized, human or chimeric antibody.
  • The antigen-binding portion of an antibody usually refers to full length or one or more fragments of an antibody that retains the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments of an antibody include a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; a F(ab)2 fragment, a bivalent fragment comprising two Fab fragments, each of which binds to the same antigen, linked by a disulfide bridge at the hinge region; a Fd fragment consisting of the VH and CH1 domains; a Fv fragment consisting of the VL and VH domains of a single arm of an antibody; and a dAb fragment, which consists of a VH domain.
  • The “Fab part” of an antibody in particular refers to a part of the antibody comprising the heavy and light chain variable regions (VH and VL) and the first domains of the heavy and light chain constant regions (CH1 and CL). In cases where the antibody does not comprise all of these regions, then the term “Fab part” only refers to those of the regions VH, VL, CH1 and CL which are present in the antibody. Preferably, “Fab part” refers to that part of an antibody corresponding to the fragment obtained by digesting a natural antibody with papain which contains the antigen binding activity of the antibody. In particular, the Fab part of an antibody encompasses the antigen binding site or antigen binding ability thereof. Preferably, the Fab part comprises at least the VH region of the antibody.
  • The “Fc part” of an antibody in particular refers to a part of the antibody comprising the heavy chain constant regions 2, 3 and—where applicable—4 (CH2, CH3 and CH4). In particular, the Fc part comprises two of each of these regions. In cases where the antibody does not comprise all of these regions, then the term “Fc part” only refers to those of the regions OH2, H3 and CH4 which are present in the antibody. Preferably, the Fc part comprises at least the CH2 region of the antibody. Preferably, “Fc part” refers to that part of an antibody corresponding to the fragment obtained by digesting a natural antibody with papain which does not contain the antigen binding activity of the antibody. In particular, the Fc part of an antibody is capable of binding to the Fc receptor and thus, e.g. comprises an Fc receptor binding site or an Fc receptor binding ability.
  • The terms “antibody” and “antibody construct”, as used herein, refer in certain embodiments to a population of antibodies or antibody constructs, respectively, of the same kind. In particular, all antibodies or antibody constructs of the population exhibit the features used for defining the antibody or antibody construct. In certain embodiments, all antibodies or antibody constructs in the population have the same amino acid sequence. Reference to a specific kind of antibody, such as an antibody capable of specifically binding to MUC1, in particular refers to a population of this kind of antibody.
  • The term “antibody” as used herein also includes fragments and derivatives of said antibody. A “fragment or derivative” of an antibody in particular is a protein or glycoprotein which is derived from said antibody and is capable of binding to the same antigen, in particular to the same epitope as the antibody. Thus, a fragment or derivative of an antibody herein generally refers to a functional fragment or derivative. In particularly preferred embodiments, the fragment or derivative of an antibody comprises a heavy chain variable region. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody or derivatives thereof. Examples of fragments of an antibody include (i) Fab fragments, monovalent fragments consisting of the variable region and the first constant domain of each the heavy and the light chain; (ii) F(ab)2 fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) Fd fragments consisting of the variable region and the first constant domain CH1 of the heavy chain; (iv) Fv fragments consisting of the heavy chain and light chain variable region of a single arm of an antibody; (v) scFv fragments, Fv fragments consisting of a single polypeptide chain; (vi) (Fv)2 fragments consisting of two Fv fragments covalently linked together; (vii) a heavy chain variable domain; and (viii) multibodies consisting of a heavy chain variable region and a light chain variable region covalently linked together in such a manner that association of the heavy chain and light chain variable regions can only occur intermolecular but not intramolecular. Derivatives of an antibody in particular include antibodies which bind to or compete with the same antigen as the parent antibody, but which have a different amino acid sequence than the parent antibody from which it is derived. These antibody fragments and derivatives are obtained using conventional techniques known to those with skill in the art.
  • A target amino acid sequence is “derived” from or “corresponds” to a reference amino acid sequence if the target amino acid sequence shares a homology or identity over its entire length with a corresponding part of the reference amino acid sequence of at least 75%, more preferably at least 80%, at least 85%, at least 90%, at least 93%, at least 95%, at least 97%, at least 98% or at least 99%. The “corresponding part” means that, for example, framework region 1 of a heavy chain variable region (FRH1) of a target antibody corresponds to framework region 1 of the heavy chain variable region of the reference antibody. In particular embodiments, a target amino acid sequence which is “derived” from or “corresponds” to a reference amino acid sequence is 100% homologous, or in particular 100% identical, over its entire length with a corresponding part of the reference amino acid sequence. A “homology” or “identity” of an amino acid sequence or nucleotide sequence is preferably determined according to the invention over the entire length of the reference sequence or over the entire length of the corresponding part of the reference sequence which corresponds to the sequence which homology or identity is defined. An antibody derived from a parent antibody which is defined by one or more amino acid sequences, such as specific CDR sequences or specific variable region sequences, in particular is an antibody having amino acid sequences, such as CDR sequences or variable region sequences, which are at least 75%, preferably at least 80%, at least 85%, at least 90%, at least 93%, at least 95%, at least 97%, at least 98% or at least 99% homologous or identical, especially identical, to the respective amino acid sequences of the parent antibody. In certain embodiments, the antibody derived from (i.e. derivative of) a parent antibody comprises the same CDR sequences as the parent antibody, but differs in the remaining sequences of the variable regions.
  • The term “antibody” as used herein also refers to multivalent and multispecific antibodies, i.e. antibody constructs which have more than two binding sites each binding to the same epitope and antibody constructs which have one or more binding sites binding to a first epitope and one or more binding sites binding to a second epitope, and optionally even further binding sites binding to further epitopes.
  • “Specific binding” preferably means that an agent such as an antibody binds stronger to a target such as an epitope for which it is specific compared to the binding to another target. An agent binds stronger to a first target compared to a second target if it binds to the first target with a dissociation constant (Kd) which is lower than the dissociation constant for the second target. Preferably the dissociation constant for the target to which the agent binds specifically is more than 100-fold, 200-fold, 500-fold or more than 1000-fold lower than the dissociation constant for the target to which the agent does not bind specifically. Furthermore, the term “specific binding” in particular indicates a binding affinity between the binding partners with an affinity constant Ka of at least 106 M−1, preferably at least 107 M−1, more preferably at least 108 M−1. An antibody specific for a certain antigen in particular refers to an antibody which is capable of binding to said antigen with an affinity having a Ka of at least 106 M−1, preferably at least 107 M−1, more preferably at least 108 M−1. For example, the term “anti-MUC1 antibody” in particular refers to an antibody specifically binding MUC1 and preferably is capable of binding to MUC1 with an affinity having a Ka of at least 106 M−1, preferably at least 107 M−1, more preferably at least 108 M−1.
  • The term “MUC1” refers to the protein MUC1, also known as mucin-1, polymorphic epithelial mucin (PEM) or cancer antigen 15-3, in particular to human MUC1 (Accession No. P15941). MUC1 is a member of the mucin family and encodes a membrane bound, glycosylated phosphoprotein. MUC1 has a core protein mass of 120-225 kDa which increases to 250-500 kDa with glycosylation. It extends 200-500 nm beyond the surface of the cell. The protein is anchored to the apical surface of many epithelial cells by a transmembrane domain. The extracellular domain includes a 20 amino acid variable number tandem repeat (VNTR) domain, with the number of repeats varying from 20 to 120 in different individuals. These repeats are rich in serine, threonine and proline residues which permits heavy O-glycosylation. In certain embodiments, the term “MUC1” refers to tumor-associated MUC1 (“TA-MUC1”). TA-MUC1 is MUC1 present on cancer cells. This MUC1 differs from MUC1 present on non-cancer cells in its much higher expression level, its localization and its glycosylation. In particular, TA-MUC1 is present apolarly over the whole cell surface in cancer cells, while in non-cancer cells MUC1 has a strictly apical expression and hence, is not accessible for systemically administered antibodies. Furthermore, TA-MUC1 has an aberrant O-glycosylation which exposes new peptide epitopes on the MUC1 protein backbone and new carbohydrate tumor antigens such as the Thomsen-Friedenreich antigen alpha (TFα).
  • “TFα”, also called Thomsen-Friedenreich antigen alpha or Core-1, refers to the disaccharide Gal-ß1,3-GalNAc which is O-glycosidically linked in an alpha-anomeric configuration to the hydroxy amino acids serine or threonine of proteins in carcinoma cells.
  • The term “sialic acid” in particular refers to any N- or O-substituted derivatives of neuraminic acid. It may refer to both 5-N-acetylneuraminic acid and 5-N-glycolylneuraminic acid, but preferably only refers to 5-N-acetylneuraminic acid. The sialic acid, in particular the 5-N-acetylneuraminic acid preferably is attached to a carbohydrate chain via a 2,3- or 2,6-linkage. Preferably, in the antibodies described herein both 2,3- as well as 2,6-coupled sialic acids are present.
  • A “relative amount of glycans” according to the invention refers to a specific percentage or percentage range of the glycans attached to the antibodies of an antibody preparation or in a composition comprising antibodies, respectively. In particular, the relative amount of glycans refers to a specific percentage or percentage range of all glycans comprised in the antibodies and thus, attached to the polypeptide chains of the antibodies in an antibody preparation or in a composition comprising antibodies. 100% of the glycans refers to all glycans attached to the antibodies of the antibody preparation or in a composition comprising antibodies, respectively. For example, a relative amount of glycans carrying bisecting GlcNAc of 10% refers to a composition comprising antibodies wherein 10% of all glycans comprised in the antibodies and thus, attached to the antibody polypeptide chains in said composition comprise a bisecting GlcNAc residue while 90% of all glycans comprised in the antibodies and thus, attached to the antibody polypeptide chains in said composition do not comprise a bisecting GlcNAc residue. The corresponding reference amount of glycans representing 100% may either be all glycan structures attached to the antibodies in the composition, or all N-glycans, i.e. all glycan structures attached to an asparagine residue of the antibodies in the composition, or all complex-type glycans. The reference group of glycan structures generally is explicitly indicated or directly derivable from the circumstances by the skilled person.
  • The term “N-glycosylation” refers to all glycans attached to asparagine residues of the polypeptide chain of a protein. These asparagine residues generally are part of N-glycosylation sites having the amino acid sequence Asn-Xaa-Ser/Thr, wherein Xaa may be any amino acid except for proline. Likewise, “N-glycans” are glycans attached to asparagine residues of a polypeptide chain. The terms “glycan”, “glycan structure”, “carbohydrate”, “carbohydrate chain” and “carbohydrate structure” are generally used synonymously herein. N-glycans generally have a common core structure consisting of two N-acetylglucosamine (GlcNAc) residues and three mannose residues, having the structure Manα1,6-(Manα1,3-)Manβ1,4-GlcNAcβ1,4-GlcNAcβ1-Asn with Asn being the asparagine residue of the polypeptide chain. N-glycans are subdivided into three different types, namely complex-type glycans, hybrid-type glycans and high mannose-type glycans.
  • The numbers given herein, in particular the relative amounts of a specific glycosylation property, are preferably to be understood as approximate numbers. In particular, the numbers preferably may be up to 10% higher and/or lower, in particular up to 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2% or 1% higher and/or lower.
  • In a “conjugate” two or more compounds are linked together. In certain embodiments, at least some of the properties from each compound are retained in the conjugate. Linking may be achieved by a covalent or non-covalent bond. Preferably, the compounds of the conjugate are linked via a covalent bond. The different compounds of a conjugate may be directly bound to each other via one or more covalent bonds between atoms of the compounds. Alternatively, the compounds may be bound to each other via a chemical moiety such as a linker molecule wherein the linker is covalently attached to atoms of the compounds. If the conjugate is composed of more than two compounds, then these compounds may, for example, be linked in a chain conformation, one compound attached to the next compound, or several compounds each may be attached to one central compound.
  • The term “nucleic acid” includes single-stranded and double-stranded nucleic acids and ribonucleic acids as well as deoxyribonucleic acids. It may comprise naturally occurring as well as synthetic nucleotides and can be naturally or synthetically modified, for example by methylation, 5′- and/or 3′-capping.
  • The term “expression cassette” in particular refers to a nucleic acid construct which is capable of enabling and regulating the expression of a coding nucleic acid sequence introduced therein. An expression cassette may comprise promoters, ribosome binding sites, enhancers and other control elements which regulate transcription of a gene or translation of an mRNA. The exact structure of expression cassette may vary as a function of the species or cell type, but generally comprises 5′-untranscribed and 5′- and 3′-untranslated sequences which are involved in initiation of transcription and translation, respectively, such as TATA box, capping sequence, CAAT sequence, and the like. More specifically, 5′-untranscribed expression control sequences comprise a promoter region which includes a promoter sequence for transcriptional control of the operatively connected nucleic acid. Expression cassettes may also comprise enhancer sequences or upstream activator sequences.
  • According to the invention, the term “promoter” refers to a nucleic acid sequence which is located upstream (5′) of the nucleic acid sequence which is to be expressed and controls expression of the sequence by providing a recognition and binding site for RNA-polymerases. The “promoter” may include further recognition and binding sites for further factors which are involved in the regulation of transcription of a gene. A promoter may control the transcription of a prokaryotic or eukaryotic gene. Furthermore, a promoter may be “inducible”, i.e. initiate transcription in response to an inducing agent, or may be “constitutive” if transcription is not controlled by an inducing agent. A gene which is under the control of an inducible promoter is not expressed or only expressed to a small extent if an inducing agent is absent. In the presence of the inducing agent the gene is switched on or the level of transcription is increased. This is mediated, in general, by binding of a specific transcription factor.
  • The term “vector” is used here in its most general meaning and comprises any intermediary vehicle for a nucleic acid which enables said nucleic acid, for example, to be introduced into prokaryotic and/or eukaryotic cells and, where appropriate, to be integrated into a genome. Vectors of this kind are preferably replicated and/or expressed in the cells. Vectors comprise plasmids, phagemids, bacteriophages or viral genomes. The term “plasmid” as used herein generally relates to a construct of extrachromosomal genetic material, usually a circular DNA duplex, which can replicate independently of chromosomal DNA.
  • According to the invention, the term “host cell” relates to any cell which can be transformed or transfected with an exogenous nucleic acid. The term “host cells” comprises according to the invention prokaryotic (e.g. E. coli) or eukaryotic cells (e.g. mammalian cells, in particular human cells, yeast cells and insect cells). Particular preference is given to mammalian cells such as cells from humans, mice, hamsters, pigs, goats, or primates. The cells may be derived from a multiplicity of tissue types and comprise primary cells and cell lines. A nucleic acid may be present in the host cell in the form of a single copy or of two or more copies and, in one embodiment, is expressed in the host cell.
  • The term “patient” means according to the invention a human being, a nonhuman primate or another animal, in particular a mammal such as a cow, horse, pig, sheep, goat, dog, cat or a rodent such as a mouse and rat. In a particularly preferred embodiment, the patient is a human being.
  • The term “cancer” according to the invention in particular comprises leukemias, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, endometrial cancer, kidney cancer, adrenal cancer, thyroid cancer, blood cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, liver cancer, colon cancer, stomach cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, pancreas cancer, ear, nose and throat (ENT) cancer, breast cancer, prostate cancer, bladder cancer, cancer of the uterus, ovarian cancer and lung cancer and the metastases thereof. The term cancer according to the invention also comprises cancer metastases.
  • By “tumor” is meant a group of cells or tissue that is formed by misregulated cellular proliferation. Tumors may show partial or complete lack of structural organization and functional coordination with the normal tissue, and usually form a distinct mass of tissue, which may be either benign or malignant.
  • By “metastasis” is meant the spread of cancer cells from its original site to another part of the body. The formation of metastasis is a very complex process and normally involves detachment of cancer cells from a primary tumor, entering the body circulation and settling down to grow within normal tissues elsewhere in the body. When tumor cells metastasize, the new tumor is called a secondary or metastatic tumor, and its cells normally resemble those in the original tumor. This means, for example, that, if breast cancer metastasizes to the lungs, the secondary tumor is made up of abnormal breast cells, not of abnormal lung cells. The tumor in the lung is then called metastatic breast cancer, not lung cancer.
  • The term “pharmaceutical composition” particularly refers to a composition suitable for administering to a human or animal, i.e., a composition containing components which are pharmaceutically acceptable. Preferably, a pharmaceutical composition comprises an active compound or a salt or prodrug thereof together with a carrier, diluent or pharmaceutical excipient such as buffer, preservative and tonicity modifier.
  • Numeric ranges described herein are inclusive of the numbers defining the range. The headings provided herein are not limitations of the various aspects or embodiments of this invention which can be read by reference to the specification as a whole. According to one embodiment, subject-matter described herein as comprising certain steps in the case of methods or as comprising certain ingredients in the case of compositions refers to subject-matter consisting of the respective steps or ingredients. It is preferred to select and combine preferred aspects and embodiments described herein and the specific subject-matter arising from a respective combination of preferred embodiments also belongs to the present disclosure.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is based on the development of a variant of the humanized anti-MUC1 antibody PankoMab wherein the glycosylation site in the CDR-H2 is deleted.
  • Deletion of the glycosylation site was achieved by substituting amino acid Asn (asparagine) 57 of the heavy chain variable region by another amino acid, especially Gln (glutamine). Asn 57 is the acceptor amino acid residue of the glycosylation site to which the carbohydrate structure is attached. Substituting this asparagine residue by another residue abolishes glycosylation because the carbohydrate structure can only be transferred to an asparagine residue by the enzymes of the host cell. It was surprisingly found that deletion of the glycosylation site in the CDR-H2 of PankoMab increased the antigen binding affinity of the antibody.
  • In view of these findings, the present invention provides an antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • Binding to MUC1
  • The antibody specifically binds to an epitope of MUC1. “Specific binding” means binding which is not non-specific adsorption. Examples of criteria for determination on whether binding is specific or not can include a dissociation constant (herein referred to as “K”). The epitope is in the extracellular tandem repeats of MUC1. In certain embodiments, the antibody binds to MUC1 in a glycosylation-dependent manner. In particular, the antibody binds stronger if said tandem repeats are glycosylated at a threonine residue with N-acetyl galactosamine (Tn), sialyl α2-6 N-acetyl galactosamine (sTn), galactose ß1-3 N-acetyl galactosamine (TF) or galactose ß1-3 (sialyl α2-6)N-acetyl galactosamine (sTF), preferably with Tn or TF. Preferably, the carbohydrate moiety is bound to the threonine residue by an α-O-glycosidic bond. The epitope in the tandem repeat domain of MUC1 in particular comprises the amino acid sequence PDTR (SEQ ID NO: 13) or PESR (SEQ ID NO: 14). The binding to this epitope preferably is glycosylation dependent, as described above, wherein in particular the binding is increased if the carbohydrate moiety described above is attached to the threonine residue of the sequence PDTR or PESR (SEQ ID NOs: 13 and 14), respectively.
  • The epitope is a tumor-associated MUC1 epitope (TA-MUC1). A TA-MUC1 epitope in particular refers to an epitope of MUC1 which is present on tumor cells but not on normal cells and/or which is only accessible by antibodies in the host's circulation when present on tumor cells but not when present on normal cells. In certain embodiments, the binding of the antibody to cells expressing TA-MUC1 epitope is stronger than the binding to cells expressing normal, non-tumor MUC1. Preferably, said binding is at least 1.5-fold stronger, preferably at least 2-fold stronger, at least 5-fold stronger, at least 10-fold stronger or at least 100-fold stronger. For TA-MUC1 binding, the antibody preferably specifically binds the glycosylated MUC1 tumor epitope such that the strength of the bond is increased at least by a factor 2, preferably a factor of 4 or a factor of 10, most preferably a factor of 20 in comparison with the bond to the non-glycosylated peptide of identical length and identical peptide sequence. Said binding can be assayed or determined by ELISA, RIA, surface plasmon resonance (hereinafter, referred to as “SPR”) analysis, or the like. Examples of equipment used in the SPR analysis can include BIAcore™ (manufactured by GE Healthcare Bio-Sciences Crop.), ProteOn™ (manufactured by Bio-Rad Laboratories, Inc.), DRX2 Biosensor (manufactured by Dynamic Biosensors GmbH), SPR-Navi™ (manufactured by BioNavis Oy Ltd.), Spreeta™ (manufactured by Texas Instruments Inc.), SPRi-Plexll™ (manufactured by Horiba, Ltd.), and Autolab SPR™ (manufactured by Metrohm). The binding of the antibody to the antigen expressed on cell surface can be assayed by flow cytometry or the like.
  • Furthermore, the antibody may exhibit antigen binding properties similar to those of a reference antibody comprising a heavy chain variable region with the amino acid sequence of SEQ ID NO: 11 or SEQ ID NO:10 and a light chain variable region with the amino acid sequence of SEQ ID NO: 12. Preferably, the reference antibody is the humanized antibody PankoMab. In particular, the antibody according to the invention specifically binds to the same antigen as the reference antibody, and preferably binds to said antigen with a higher affinity. That is, the antibody preferably binds to the antigen with an affinity having a dissociation constant which is lower than that of the reference antibody, more preferably at least 10% lower, at least 20% lower, at least 30% lower or at least 50% lower. Moreover, the antibody preferably shows cross-specificity with the reference antibody comprising a heavy chain variable region with the amino acid sequence of SEQ ID NO: 11 and a light chain variable region with the amino acid sequence of SEQ ID NO: 12. In particular, the humanized antibody is able to block the binding of the reference antibody to MUC1 if present in a high enough concentration. This is possible if the binding of the reference antibody to MUC1 is hindered when the antibody according to the invention is already bound to the antigen MUC1.
  • The Anti-MUC1 Antibody
  • An antibody capable of binding to MUC1 of the present invention comprises a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • In certain embodiments, the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 9. Especially, the heavy chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 9. In these embodiments, the heavy chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 1, 2 and 3. Hence, any sequence deviations to SEQ ID NO: 9 are located in the framework regions, but not in the CDRs. In particular, the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 9.
  • In certain embodiments, CDR-H2 has the amino acid sequence of SEQ ID NO: 2, wherein the amino acid at position 8 of SEQ ID NO: 2 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine; especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine. Preferably, the amino acid at position 8 of SEQ ID NO: 2 is glutamine, histidine, tryptophan, lysine or arginine, especially glutamine. In particular, CDR-H2 has the amino acid sequence of SEQ ID NO: 7.
  • In specific embodiments, the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 10. Especially, the heavy chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 10. In these embodiments, the heavy chain variable region comprises the CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 7 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3. Hence, any sequence deviations to SEQ ID NO: 10 are located in the framework regions, but not in the CDRs. In particular, the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 10.
  • In certain embodiments, the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12. Especially, the light chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 12. In these embodiments, the light chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 4, 5 and 6. Hence, any sequence deviations to SEQ ID NO: 12 are located in the framework regions, but not in the CDRs. In particular, the light chain variable region comprises the amino acid sequence of SEQ ID NO: 12.
  • In specific embodiments, the heavy chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 9, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 2 and 3, and the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6. In particular, the heavy chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 9, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 2 and 3, and the light chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • In specific embodiments, the heavy chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 10, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3, and the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6. In particular, the heavy chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 10, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3, and the light chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • In specific embodiments, the heavy chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20. Especially, the heavy chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20. In these embodiments, the heavy chain variable region comprises the CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3. Hence, any sequence deviations to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20 are located in the framework regions, but not in the CDRs. In particular, the heavy chain variable region comprises the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20. In certain embodiments, the amino acid at position 76 of SEQ ID NO: 20 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine; especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine.
  • Preferably, the amino acid at position 76 of SEQ ID NO: 20 is glutamine, histidine, tryptophan, lysine or arginine, especially glutamine. In particular, CDR-H2 has the amino acid sequence of SEQ ID NO: 7 and/or the heavy chain variable region comprises the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 23.
  • In specific embodiments, the light chain variable region comprises an amino acid sequence which is at least 90% identical to the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21. Especially, the light chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21. In these embodiments, the light chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 4, 5 and 6. Hence, any sequence deviations to amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21 are located in the framework regions, but not in the CDRs. In particular, the light chain variable region comprises the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21.
  • In specific embodiments, the heavy chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3, and the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6. In particular, the heavy chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3, and the light chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • In specific embodiments, the heavy chain comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 15. Especially, the heavy chain comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 15. In these embodiments, the heavy chain comprises the CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3. Hence, any sequence deviations to SEQ ID NO: 15 are located in the framework regions, but not in the CDRs. In particular, the heavy chain comprises the amino acid sequence of SEQ ID NO: 15. In certain embodiments, the amino acid at position 57 of SEQ ID NO: 15 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine; especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine. Preferably, the amino acid at position 57 of SEQ ID NO: 15 is glutamine, histidine, tryptophan, lysine or arginine, especially glutamine. In particular, CDR-H2 has the amino acid sequence of SEQ ID NO: 7 and/or the heavy chain variable region comprises the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 22.
  • In specific embodiments, the light chain comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 16. Especially, the light chain comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 16. In these embodiments, the light chain still comprises CDRs having the amino acid sequences of SEQ ID NOs: 4, 5 and 6. Hence, any sequence deviations to SEQ ID NO: 16 are located in the framework regions, but not in the CDRs. In particular, the light chain comprises the amino acid sequence of SEQ ID NO: 16.
  • In specific embodiments, the heavy chain has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 15, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3, and the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 16, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6. In particular, the heavy chain has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 15, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 7 and 3, and the light chain has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 16, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • The antibody according to the present invention includes and encompasses modified forms thereof. The modified form of the antibody of the present invention means an antibody of the present invention provided with chemical or biological modification. The chemically modified form includes forms wherein the amino acid skeleton is conjugated with a chemical moiety, forms having a chemically modified N-linked or O-linked carbohydrate chain, and the like. Said chemical moiety or form can be toxic or cytotoxic. Biologically modified forms include forms that have undergone post-translational modification (e.g., N-linked or O-linked glycosylation, N-terminal or C-terminal processing, deamidation, isomerization of aspartic acid, or oxidation of methionine), forms containing a methionine residue added to the N-terminus by expression using prokaryotic host cells, and the like. Such a modified form is also meant to include a form labeled to permit detection or isolation of the antibody of the present invention or the antigen thereof, for example, enzyme-labeled forms, fluorescently labeled forms, and affinity-labeled forms. Such a modified form of the antibody of the present invention is useful in improvement in the stability or blood retention of the original antibody of the present invention, reduction in antigenicity, detection or isolation of the antibody or the antigen, etc.
  • In particular, the antibody may comprise one or more modifications selected from the group consisting of defucosylation, reduced fucose, N-linked glycosylation, O-linked glycosylation, N-terminal processing, C-terminal processing, deamidation, isomerization of aspartic acid, oxidation of methionine, substitutions of two leucine (L) residues to alanine (A) at position 234 and 235 of the heavy chain (LALA), amidation of a proline residue and deletion or lack of one, two, or three amino acids at the carboxyl terminus. In specific embodiments, the antibody lacks one, two, or three carboxyl-terminal amino acid(s) at one or both heavy chains, or it lacks one carboxyl-terminal amino acid and the carboxyl-terminal proline residues is amidated at one or both heavy chains.
  • Such a modification may be made at an arbitrary position or the desired position in the antibody thereof. Alternatively, the same or two or more different modifications may be made at one or two or more positions therein.
  • For example, antibodies produced by cultured mammalian cells are known to lack a carboxyl-terminal lysine residue in its heavy chain (Journal of Chromatography A, 705: 129-134 (1995)). It is also known that occasionally 2 carboxyl-terminal amino acid residues (i.e., glycine and lysine) of a heavy chain are missing and that a proline residue newly located at the carboxyl terminus is amidated (Analytical Biochemistry, 360: 75-83 (2007)). Such lack or modification in these heavy chain sequences, however, affects neither the ability of the antibody to bind to its antigen nor the effector functions (complement activation, antibody-dependent cytotoxicity, etc.) of the antibody.
  • In certain embodiments, the antibody comprises a deletion or lack of 1 or 2 amino acid(s) in the carboxyl terminus of the heavy chain, and has an amidated residue (e.g., an amidated proline residue at the carboxyl-terminal site of the heavy chain). However, the antibody according to the present invention is not limited to the types described above as long as the deletion mutant maintains the ability to bind to the antigen.
  • In certain embodiments, two heavy chains of the antibody according to the present invention may be composed of any one type of heavy chain selected from the group consisting of the full length heavy chains and the heavy chains of the deletion mutant or may be composed of the combination of any two types selected therefrom. The quantitative ratio of the deletion variant heavy chain(s) depends on the type of cultured mammalian cells producing the antibody according to the present invention, and the culture conditions of the cells.
  • In specific embodiments, the antibody according to the present invention can include two heavy chains, both of which lack one carboxyl-terminal amino acid residue.
  • In specific embodiments, the antibody comprises the heavy chain having an amino acid sequence represented by amino acid Nos 1 to 446 of SEQ ID NO: 15 or 22 and the light chain having an amino acid sequence represented by amino acid Nos 1 to 219 of SEQ ID NO: 16. In certain embodiments, the amino acid at position 57 of SEQ ID NO: 15 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine; especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine. Preferably, the amino acid at position 57 of SEQ ID NO: 15 is glutamine, histidine, tryptophan, lysine or arginine, especially glutamine.
  • In certain embodiments, the antibody according to the present invention competes for the binding to TA-MUC1 with an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region having the amino acid sequence of SEQ ID NO: 12, or an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 11 and a light chain variable region having the amino acid sequence of SEQ ID NO: 12.
  • In certain embodiments, the antibody has the following properties: (a) specifically binding to MUC1, and/or (b) having the activity of being internalized into MUC1-expressing cells through binding to MUC1.
  • In certain embodiments, the antibody comprises at least one antibody heavy chain. Especially, the antibody comprises two antibody heavy chains. The antibody heavy chains in particular comprise a VH domain, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain. In certain other embodiments, the antibody heavy chains comprise a CH2 domain and a CH3 domain, but do not comprise a CH1 domain. In further embodiments, one or more constant domains of the heavy chains may be replaced by other domains, in particular similar domains such as for example albumin. The antibody heavy chains may be of any type, including γ-, α-, ε-, δ- and μ-chains, and preferably are γ-chains, including γ1-, γ2-, γ3- and γ4-chains, especially γ1-chains. Hence, the antibody preferably is an IgG-type antibody such as an IgG1-, IgG3- or IgG4-type antibody, in particular an IgG1-type antibody.
  • In particular, the antibody further comprises at least one antibody light chain, especially two antibody light chains. The antibody light chains in particular comprise a VL domain and a CL domain. The antibody light chain may be a κ-chain or a λ-chain and especially is a κ-chain.
  • In certain embodiments, the antibody comprises two antibody heavy chains and two antibody light chains. In particular, the antibody comprises two antibody heavy chains of the γ1-type, each comprising a VH domain, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain, and two antibody light chains of the κ-type, each comprising a VL domain and a CL domain.
  • In alternative embodiments, the antibody does not comprise an antibody light chain. In these embodiments, the light chain variable region may be fused to the N terminus of the heavy chain variable region or is inserted C terminal to the heavy chain variable region. Peptide linkers may be present to connect the light chain variable region with the remaining parts of the heavy chain.
  • In preferred embodiments, the antibody comprises an Fc region. The antibody may especially be a whole antibody, comprising two heavy chains each comprising the domains VH, CH1, hinge region, CH2 and CH3, and two light chains each comprising the domains VL and CL. The antibody in particular is capable of binding to one or more human Fcγ receptors, especially human Fcγ receptor IIIA. In alternative embodiments, the antibody does not or not significantly bind the human Fcγ receptor IIIA, and especially does not or not significantly bind to any human Fcγ receptor. In these embodiments the antibody in particular does not comprise a glycosylation site in the CH2 domain.
  • In alternative embodiments, the antibody does not comprise an Fc region. In these embodiments, the antibody in particular is a single chain variable region fragment (scFv) or another antibody fragment not comprising an Fc region.
  • Glycosylation of the Anti-MUC1 Antibody
  • The anti-MUC1 antibody may comprise a CH2 domain in one or more antibody heavy chains. Natural human antibodies of the IgG type comprise an N-glycosylation site in the CH2 domain. The CH2 domains present in the antibody may or may not comprise an N-glycosylation site. In certain embodiments, the antibody does not comprise a glycosylation site in the CH2 domain. In particular, the antibody does not comprise an asparagine residue at the position in the heavy chain corresponding to position 297 according to the IMGT/Eu numbering system. For example, the antibody may comprise an Ala297 mutation in the heavy chain. In these embodiments, the antibody preferably has a strongly reduced ability or completely lacks the ability to induce, via binding to Fcγ receptors, antibody-dependent cellular cytotoxicity (ADCC) and/or antibody-dependent cellular phagocytosis (ADCP) and/or complement-dependent cytotoxicity (CDC). Strongly reduced ability in this respect in particular refers to a reduction to 10% or less, especially 3% or less, 1% or less or 0.1% or less activity compared to the same antibody comprising an N-glycosylation site in its CH2 domains and having a common mammalian glycosylation pattern such as those obtainable by production in human cell lines or in CHO cell lines, for example a glycosylation pattern as described herein. In these embodiments, the antibody in particular is an IgG1-type antibody.
  • In alternative embodiments, the CH2 domains present in the antibody comprise an N-glycosylation site. This glycosylation site in particular is at an amino acid position corresponding to amino acid position 297 of the heavy chain according to the IMGT/Eu numbering system and has the amino acid sequence motive Asn Xaa Ser/Thr wherein Xaa may be any amino acid except proline. The N-linked glycosylation at Asn297 is conserved in mammalian IgGs as well as in homologous regions of other antibody isotypes. Due to optional additional amino acids which may be present in the variable region or other sequence modifications, the actual position of this conserved glycosylation site may vary in the amino acid sequence of the antibody. Preferably, the glycans attached to the antibody are biantennary complex type N-linked carbohydrate structures, preferably comprising at least the following structure:

  • Asn-GlcNAc-GlcNAc-Man-(Man-GlcNAc)2
  • wherein Asn is the asparagine residue of the polypeptide portion of the antibody; GlcNAc is N-acetylglucosamine and Man is mannose. The terminal GlcNAc residues may further carry a galactose residue, which optionally may carry a sialic acid residue. A further GlcNAc residue (named bisecting GlcNAc) may be attached to the Man nearest to the polypeptide. A fucose may be bound to the GlcNAc attached to the Asn. In these embodiments, the antibody in particular is an IgG1-type antibody.
  • In preferred embodiments, the antibody does not comprise N-glycolyl neuraminic acids (NeuGc) or detectable amounts of NeuGc. Furthermore, the antibody preferably also does not comprise Galili epitopes (Galα1,3-Gal structures) or detectable amounts of the Galili epitope. In particular, the relative amount of glycans carrying NeuGc and/or Galα1,3-Gal structures is less than 0.1% or even less than 0.02% of the total amount of glycans attached to the CH2 domains of the antibodies in the population of antibodies.
  • In particular, the antibody has a human glycosylation pattern. Due to these glycosylation properties, foreign immunogenic non-human structures which induce side effects are absent which means that unwanted side effects or disadvantages known to be caused by certain foreign sugar structures such as the immunogenic non-human sialic acids (NeuGc) or the Galili epitope (Gal-Gal structures), both known for rodent production systems, or other structures like immunogenic high-mannose structures as known from e.g. yeast systems are avoided.
  • In specific embodiments, the antibody comprises a glycosylation pattern having a detectable amount of glycans carrying a bisecting GlcNAc residue. In particular, the relative amount of glycans carrying a bisecting GlcNAc residue is at least 0.5%, especially at least 1% of the total amount of glycans attached to the glycosylation sites of the antibody in a composition. Furthermore, in certain embodiments the glycosylation pattern comprises a relative amount of glycans carrying at least one galactose residue of at least 25% of the total amount of glycans attached to the antibody in a composition. In particular, the relative amount of glycans carrying at least one galactose residue is at least 30%, especially at least 35% or at least 40% of the total amount of glycans attached to the antibody in a composition. In specific embodiments, the glycosylation pattern comprises a relative amount of glycans carrying at least one sialic acid residue of at least 1% of the total amount of glycans attached to the antibody in a composition. In particular, the relative amount of glycans carrying at least one sialic acid residue is at least 1.5%, especially at least 2% of the total amount of glycans attached to the antibody in a composition.
  • The antibody may have a glycosylation pattern having a high amount of core fucose or a low amount of core fucose. A reduced amount of fucosylation increases the ability of the antibody to induce ADCC. In certain embodiments, the relative amount of glycans carrying a core fucose residue is 40% or less, especially 30% or less or 20% or less of the total amount of glycans attached to the antibody in a composition. In alternative embodiments, the relative amount of glycans carrying a core fucose residue is at least 60%, especially at least 65% or at least 70% of the total amount of glycans attached to the antibody in a composition.
  • Via the presence or absence of the glycosylation site in the CH2 domain of the anti-MUC1 antibody and the presence or absence of fucose in the glycan structures at said glycosylation site, the ability of the antibody to induce ADCC and the strength of said ADCC induction can be controlled. The ADCC activity is increased by glycosylation of the Fc part of the antibody and further by reducing the amount of fucosylation in said glycosylation. In certain applications, fine tuning of the ADCC activity is important.
  • Therefore, in certain situations, the antibody without a glycosylation site in the CH2 domain, the antibody with a glycosylation site in the CH2 domain and with a high amount of fucosylation, or the antibody with a glycosylation site in the CH2 domain and with a low amount of fucosylation may be most advantageous.
  • Production of the Anti-MUC1 Antibody
  • The antibody is preferably recombinantly produced in a host cell. The host cell used for the production of the antibody may be any host cells which can be used for antibody production. Suitable host cells are in particular eukaryotic host cells, especially mammalian host cells. Exemplary host cells include yeast cells such as Pichia pastoris cell lines, insect cells such as SF9 and SF21 cell lines, plant cells, bird cells such as EB66 duck cell lines, rodent cells such as CHO, NS0, SP2/0 and YB2/0 cell lines, and human cells such as HEK293, PER.C6, CAP, CAP-T, AGE1.HN, Mutz-3 and KG1 cell lines.
  • In certain embodiments, the antibody is produced recombinantly in a human blood cell line, in particular in a human myeloid leukemia cell line. Preferred human cell lines which can be used for production of the antibody as well as suitable production procedures are described in WO 2008/028686 A2. In a specific embodiment, the antibody is obtained by expression in a human myeloid leukemia cell line selected from the group consisting of NM-H9D8, NM-H9D8-E6 and NM-H9D8-E6Q12 and cell lines derived therefrom. These cell lines were deposited under the accession numbers DSM ACC2806 (NM-H9D8; deposited on Sep. 15, 2006), DSM ACC2807 (NM-H9D8-E6; deposited on Oct. 5, 2006) and DSM ACC2856 (NM-H9D8-E6Q12; deposited on Aug. 8, 2007) according to the requirements of the Budapest Treaty at the Deutsche Sammlung von Mikroorganismen und Zellkulturen (DSMZ), Inhoffenstraße 7B, 38124 Braunschweig (DE) by Glycotope GmbH, Robert-Rössle-Str. 10, 13125 Berlin (DE). NM-H9D8 cells provide a glycosylation pattern with a high degree of sialylation, a high degree of bisecting GlycNAc, a high degree of galactosylation and a high degree of fucosylation. NM-H9D8-E6 and NM-H9D8-E6Q12 cells provide a glycosylation pattern similar to that of NM-H9D8 cells, except that the degree of fucosylation is very low. Other suitable cell lines include K562, a human myeloid leukemia cell line present in the American Type Culture Collection (ATCC CCL-243), as well as cell lines derived from the aforementioned.
  • In further embodiments, the antibody is produced recombinantly in CHO cells. Especially, the antibody may be produced recombinantly in a CHO dhfr- cell line such as the cell line of ATCC No. CRL-9096.
  • Conjugates of the Anti-MUC1 Antibody
  • In certain embodiments, the antibody comprises one or more further agents conjugated thereto. The further agent may be any agent suitable for conjugation to the antibody. If more than one further agent is present in the antibody, these further agents may be identical or different, and in particular are all identical. Conjugation of the further agent to the antibody can be achieved using any methods known in the art. The further agent may be covalently, in particular by fusion or chemical coupling, or non-covalently attached to the antibody. In certain embodiments, the further agent is covalently attached to the antibody, especially via a linker moiety. The linker moiety may be any chemical entity suitable for attaching the further agent to the antibody.
  • The further agent preferably is useful in therapy, diagnosis, prognosis, detecting and/or monitoring of a disease, in particular cancer. For example, the further agent may be selected from the group consisting of radionuclides, chemotherapeutic agents, antibodies or antibody fragments, in particular those of different specificity than the anti-MUC1 antibody, e.g. checkpoint antibodies which block or activate immunomodulatory targets, enzymes, interaction domains, detectable labels, toxins, cytolytic components, immunomodulators, immunoeffectors, MHC class I or class II antigens, and liposomes. A particular preferred further agent is a radionuclide or a cytotoxic agent capable of killing cancer cells, such as a chemotherapeutic agent. In certain preferred embodiments, a chemotherapeutic agent is attached to the anti-MUC1 antibody forming a conjugate.
  • Specific examples of chemotherapeutic agents that can be conjugated as further agent include alkylating agents such as cisplatin, anti-metabolites, plant alkaloids and terpenoids, vinca alkaloids, podophyllotoxin, taxanes such as taxol, topoisomerase inhibitors such as irinotecan and topotecan, antineoplastics such as doxorubicin or microtubule inhibitors such as auristatins and maytansin/maytansinoids.
  • The chemotherapeutic agent may in particular be selected from a group consisting of a V-ATPase inhibitor, a pro-apoptotic agent, a Bcl2 inhibitor, an MCL1 inhibitor, a HSP90 inhibitor, an IAP inhibitor, an mTor inhibitor, a microtubule stabilizer, a microtubule destabilizer, an auristatin, a dolastatin, a maytansin, a maytansinoid, amatoxin, a methionine aminopeptidase, an inhibitor of nuclear export of proteins CRM1, a DPPIV inhibitor, proteasome inhibitors, inhibitors of phosphoryl transfer reactions in mitochondria, a protein synthesis inhibitor, a kinase inhibitor, a CDK2 inhibitor, a CDK9 inhibitor, a kinesin inhibitor, an HDAC inhibitor, a DNA damaging agent, a DNA alkylating agent, a DNA intercalator, a DNA minor groove binder, a DHFR inhibitor, an inhibitor of microtubule formation, a stabilizer of microtubule, a stabilizer of actin, a topoisomerase II inhibitor, a platinum compound, a ribosome inhibitor, an RNA polymerase II inhibitor and a bacterial toxin. In specific embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is selected from the group consisting of an auristatin, a microtubule inhibitor such as maytansinoid, a DNA damaging agent, a DNA alkylating agent and a DNA minor groove binder.
  • In some embodiments of the chemotherapeutic agent is a maytansin or maytansinoid. Specific examples of maytansinoids useful for conjugation include maytansinol, N2′-deacetyl-N2′-(3-mercapto-1-oxopropyl)-maytansine (DM1), N2′-deacetyl-N2′-(4-mercapto-1-oxopentyl)-maytansine (DM3), and N2′-deacetyl-N2′-(4-methyl-4-mercapto-1-oxopentyl)-maytansine (DM4). In particular, DM1 or DM4 is attached to the anti-MUC1 antibody. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is an auristatin, in particular monomethyl auristatin F (MMAF), monomethyl auristatin E (MMAE) or auristatin T. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a DNA minor groove binder, in particular pyrrolobenzodiazepine (PBD), pyrrolobenzodiazepine dimer (PBD dimer), duocarmycin, duocarmycin-hydroxybenzamide-azaindole (DUBA), seco-duocarmycin-hydroxybenzamide-azaindole (seco-DUBA) or doxorubicin. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a DNA alkylating agent, in particular indolinobenzodiazepine or oxazolidinobenzodiazepine. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a DNA damaging agent, in particular calicheamicin. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is an inhibitor of microtubule formation, in particular a tubulysin, an ansamitocin, podophyllotoxin or vinblastine. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a stabilizer of microtubuli, in particular paclitaxel or an epothilone. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a stabilizer of actin, in particular a phallotoxin. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a topoisomerase II inhibitor, in particular teniposide, XK469, razoxane, amsacrine, idarubicin or mebarone. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a platinum compound, in particular cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin or sattraplatin. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a ribosome inhibitor, in particular ricin, saporin, abrin, diphtheria toxin or exotoxin A. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is an RNA polymerase II inhibitor, in particular an amatoxin, such as, for example, amanitin. In some embodiments, the chemotherapeutic agent attached to the anti-MUC1 antibody is a bacterial toxin, in particular anthrax toxin. Suitable antibody drug conjugates are also described in EP 16 151 774.3 and LU 92659, to which is explicitly referred to herewith.
  • In certain embodiments, the further agent is a polypeptide or protein. This polypeptide or protein may in particular be fused to a polypeptide chain of the antibody. In certain embodiments, the further agent being a polypeptide or protein is fused to the C terminus of an antibody light chain of the antibody. In embodiments wherein the antibody comprises two antibody light chains, a further agent being a polypeptide or protein may be fused to the C terminus of each of the two antibody light chains. In further embodiments, the further agent being a polypeptide or protein is fused to the C terminus of an antibody heavy chain of the antibody. In embodiments wherein the antibody comprises two antibody heavy chains, a further agent being a polypeptide or protein may be fused to the C terminus of each of the two antibody heavy chains. The further agents may be identical or different and in particular have the same amino acid sequence. Suitable examples of such further agents being a polypeptide or protein may be selected from the group consisting of cytokines, chemokines, antibodies, antigen binding fragments, enzymes, and interaction domains.
  • In certain embodiments, the further agent being a polypeptide or protein is a checkpoint antibody which blocks and/or triggers activating signals. Examples of respective targets include CD40, CD3, CD137 (4-1BB), OX40, GITR, CD27, CD278 (ICOS), CD154 (CD40 ligand), CD270 (HVEM) and CD258 (LIGHT) as activating targets, CTLA4, PD1, CD80, CD244, A2AR, B7-H3 (CD276), B7-H4 (VTCN1), BTLA, IDO, KIR, LAG3, TIM-3, VISTA and phosphatidylserine as inhibitory targets, and their respective ligands such as PDL1. In specific examples, the anti-MUC1 antibody comprises two heavy chains and two light chains as described herein, wherein a scFv fragment specifically binding to CD3 is fused to the C terminus of each heavy chain; or wherein a scFv fragment specifically binding to PDL1 is fused to the C terminus of each light chain.
  • In further embodiments, the further agent being a polypeptide or protein is an immunomodulatory compound such as a chemokine, cytokine or growth factor. Suitable cytokines in this respect include interferons such as interferon-a, interferon-p and interferon-y, and interleukins. Suitable growth factors include G-CSF and GM-CSF.
  • The Nucleic Acid, Expression Cassette, Vector, Cell Line and Composition
  • In a further aspect, the present invention provides a nucleic acid encoding the antibody. The nucleic acid sequence of said nucleic acid may have any nucleotide sequence suitable for encoding the antibody. However, preferably the nucleic acid sequence is at least partially adapted to the specific codon usage of the host cell or organism in which the nucleic acid is to be expressed, in particular the human codon usage. The nucleic acid may be double-stranded or single-stranded DNA or RNA, preferably double-stranded DNA such as cDNA or single-stranded RNA such as mRNA. It may be one consecutive nucleic acid molecule or it may be composed of several nucleic acid molecules, each coding for a different part of the antibody. In preferred embodiments, the present invention provides a nucleotide sequence of the heavy chain of PankoMab variant (PM-N54Q) represented by SEQ ID NO: 17 and a nucleotide sequence of the light chain of PankoMab variant (PM-N54Q) represented by SEQ ID NO: 18.
  • If the antibody is composed of more than one different amino acid chain, such as a light chain and a heavy chain of the antibody, the nucleic acid may, for example, be a single nucleic acid molecule containing several coding regions each coding for one of the amino acid chains of the antibody, preferably separated by regulatory elements such as IRES elements in order to generate separate amino acid chains, or the nucleic acid may be composed of several nucleic acid molecules wherein each nucleic acid molecule comprises one or more coding regions each coding for one of the amino acid chains of the antibody. In addition to the coding regions encoding the antibody, the nucleic acid may also comprise further nucleic acid sequences or other modifications which, for example, may code for other proteins, may influence the transcription and/or translation of the coding region(s), may influence the stability or other physical or chemical properties of the nucleic acid, or may have no function at all.
  • In a further aspect, the present invention provides an expression cassette or vector comprising a nucleic acid according to the invention and a promoter operatively connected with said nucleic acid. In addition, the expression cassette or vector may comprise further elements, in particular elements which are capable of influencing and/or regulating the transcription and/or translation of the nucleic acid, the amplification and/or reproduction of the expression cassette or vector, the integration of the expression cassette or vector into the genome of a host cell, and/or the copy number of the expression cassette or vector in a host cell. Suitable expression cassettes and vectors comprising respective expression cassettes for expressing antibodies are well known in the prior art and thus, need no further description here.
  • Furthermore, the present invention provides a host cell comprising the nucleic acid according to the invention or the expression cassette or vector according to the invention. The host cell may be any host cell. It may be an isolated cell or a cell comprised in a tissue. Preferably, the host cell is a cultured cell, in particular a primary cell or a cell of an established cell line, preferably a tumor-derived cell. Preferably, it is a bacterial cell such as E. coli, a yeast cell such as a Saccharomyces cell, in particular S. cerevisiae, an insect cell such as a Sf9 cell, or a mammalian cell, in particular a human cell such as a tumor-derived human cell, a hamster cell such as CHO, or a primate cell. In a preferred embodiment of the invention the host cell is derived from human myeloid leukaemia cells. Preferably, it is selected from the following cells or cell lines: K562, KG1, MUTZ-3, or a cell or cell line derived therefrom, or a mixture of cells or cell lines comprising at least one of those aforementioned cells. The host cell is preferably selected from the group consisting of NM-H9D8, NM-H9D8-E6, NM H9D8-E6012, and a cell or cell line derived from anyone of said host cells. These cell lines and their properties are described in detail in the PCT-application WO 2008/028686 A2. In further embodiments, the host cell is of a CHO dhfr- cell line such as the cell line of ATCC No. CRL-9096. In preferred embodiments, the host cell is optimized for expression of glycoproteins, in particular antibodies, having a specific glycosylation pattern. Preferably, the codon usage in the coding region of the nucleic acid according to the invention and/or the promoter and the further elements of the expression cassette or vector are compatible with and, more preferably, optimized for the type of host cell used. Preferably, the antibody is produced by a host cell or cell line as described above.
  • In a specific aspect, the present invention provides a method of producing the antibody in a host cells as described herein. The method in particular comprises the steps of providing a host cell comprising a nucleic acid encoding the antibody, culturing the host cell under conditions suitable for expression of the antibody, and obtaining the antibody expressed by the host cell. The antibody according to the invention may be obtained or obtainable by said method.
  • In another aspect, the present invention provides a composition comprising the antibody, the nucleic acid, the expression cassette or vector, or the host cell. The composition may also contain more than one of these components. Furthermore, the composition may comprise one or more further components selected from the group consisting of solvents, diluents, and excipients Preferably, the composition is a pharmaceutical composition. In this embodiment, the components of the composition preferably are all pharmaceutically acceptable. The composition may be a solid or fluid composition, in particular a—preferably aqueous—solution, emulsion or suspension or a lyophilized powder.
  • Use in Medicine
  • The antibody in particular is useful in medicine, in particular in therapy, diagnosis, prognosis, detecting and/or monitoring of a disease, in particular a disease as described herein, preferably cancer, infections, inflammatory diseases, graft-versus-host disease and immunodeficiencies.
  • Therefore, in a further aspect, the invention provides the antibody, the nucleic acid, the expression cassette or vector, the host cell, or the composition for use in medicine. Preferably, the use in medicine is a use in the treatment, prognosis, diagnosis, detecting and/or monitoring of a disease such as, for example, diseases associated with abnormal cell growth such as cancer, infections such as bacterial, viral, fungal or parasitic infections, inflammatory diseases such as autoimmune diseases and inflammatory bowel diseases, and diseases associated with a reduce immune activity such as immunodeficiencies. In a preferred embodiment, the disease is cancer.
  • Preferably, the cancer has a detectable expression of MUC1 or TA-MUC1, preferably detectable by immunohistochemistry, ELISA, RIA, enzyme-linked immunospot (ELISPOT) assay, dot blotting, Ouchterlony test or counter-immunoelectrophoresis (CIE), or in-situ hybridization. It especially includes cells having an MUC1 or TA-MUC1 expression which is detectable by immunohistochemistry or in-situ hybridization. The cancer may be tested on MUC1 or TA-MUC1 level prior to administration of the anti-MUC1 antibody.
  • The present invention further provides kits and devices comprising the antibody according to the invention, and associated methods that are useful in the diagnosis, detecting or monitoring of MUC1 associated disorders such as cancer. In some embodiments, a sandwich ELISA kit for testing, detecting or diagnosis comprising the antibody of the present invention is provided. This kit may further comprise one or more of a solution of MUC1 or TA-MUC1 protein standards, a coloring reagent, a buffer solution for dilution, an antibody for solid phase, an antibody for detection, and a washing solution, and the like. Preferably, the amount of the antibody bound to the antigen can be measured by the application of a method such as an absorbance, fluorescence, luminescence, or radioisotope (RI) method. Preferably, an absorbance plate reader, a fluorescence plate reader, a luminescence plate reader, an RI liquid scintillation counter, or the like is used in the measurement.
  • In some embodiments, the present invention provides the antibody according to the invention for use in immunohistochemistry (IHC) analysis, and a composition comprising the same.
  • The immunohistochemistry is not particularly limited as long as this approach involves reacting a tissue section with an antigen-binding antibody (primary antibody) and detecting the primary antibody bound with the antigen.
  • Different forms of cancers including metastases can be treated with the antibody according to the invention. The cancer can in particular be selected from the group consisting of colon cancer, lung cancer, ovarian cancer, breast cancer such as triple negative breast cancer, pancreatic cancer, cervical cancer, endometrial cancer, gastrointestinal cancer, kidney cancer, head and neck cancer, thyroid cancer and urothelial cancer. The cancer may further in particular be selected from stomach cancer, liver cancer, bladder cancer, skin cancer, prostate cancer and blood cancer. In certain embodiments, the cancer is a metastasizing cancer. The cancer may include any type of metastases, such as skin metastases, lymph node metastases, lung metastases, liver metastases, peritoneal metastases, pleural metastases and/or brain metastases. In certain embodiments, the cancer has an inflammatory phenotype. In these embodiments, any of the cancer types described above may be an inflammatory cancer.
  • In certain embodiments, the viral infection is caused by human immunodeficiency virus, herpes simplex virus, Epstein Barr virus, influenza virus, lymphocytic choriomeningitis virus, hepatitis B virus or hepatitis C virus. The inflammatory disease may be selected from inflammatory bowel disease, pelvic inflammatory disease, ischemic stroke, Alzheimer's disease, asthma, pemphigus vulgaris and dermatitis/eczema. The autoimmune disease may be selected from the group consisting of celiac disease, diabetes mellitus type 1, Graves' disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis, systemic lupus erythematosus, vitiligo, psoriatic arthritis, atopic dermatitis, scleroderma, sarcoidosis, primary biliary cirrhosis, Guillain-Barre syndrome, autoimmune hepatitis and ankylosing spondylitis. In certain embodiments, the disease comprises or is associated with cells which express MUC1, especially TA-MUC1. For example, a cancer to be treated is MUC1 positive, especially TA-MUC1 positive, i.e. comprises cancer cells which express MUC1, especially TA-MUC1.
  • In specific embodiments, the antibody is used for treatment in combination with another therapeutic agent, especially for treatment of cancer in combination with another anti-cancer agent. Said further therapeutic agent may be any known anti-cancer agent. Suitable anti-cancer therapeutic agents which may be combined with the antibody according to the invention may be chemotherapeutic agents, other antibodies, immunostimulatory agents, cytokines, chemokines, and vaccines. Furthermore, therapy with the antibody may be combined with radiation therapy, surgery and/or traditional Chinese medicine.
  • Anti-cancer agents that can be used in combination with the anti-MUC1 antibody may be selected from any chemotherapeutic agent, in particular chemotherapeutic agents known to be effective for treatment of MUC1 positive cancers. The type of chemotherapeutic agent also depends on the cancer to be treated. The combination partner may be selected from the group consisting of taxanes such as paclitaxel (Taxol), docetaxel (Taxotere) and SB-T-1214; cyclophosphamide; imatinib; pazopanib; capecitabine; cytarabine; vinorelbine; gemcitabine; anthracyclines such as daunorubicin, doxorubicin, epirubicin, idarubicin, valrubicin and mitoxantrone; aromatase inhibitors such as aminoglutethimide, testolactone (Teslac), anastrozole (Arimidex), letrozole (Femara), exemestane (Aromasin), vorozole (Rivizor), formestane (Lentaron), fadrozole (Afema), 4-hydroxyandrostenedione, 1,4,6-androstatrien-3,17-dione (ATD) and 4-androstene-3,6,17-trione (6-OXO); topoisomerase inhibitors such as irinotecan, topotecan, camptothecin, lamellarin D, etoposide (VP-16), teniposide, doxorubicin, daunorubicin, mitoxantrone, amsacrine, ellipticines, aurintricarboxylic acid and HU-331; platinum based chemotherapeutic agents such as cis-diamminedichloroplatinum(II) (cisplatin), cis-diammine(1,1-cyclobutanedicarboxylato)platinum(II) (carboplatin) and [(1R,2R)-cyclohexane-1,2-diamine](ethanedioato-O,O′)platinum(II) (oxaliplatin); PARP inhibitors such as olaparib, rucaparib and niraparib; TLR agonists such as imiquimod and resiquimod; and antimetabolites, in particular antifolates such as methotrexate, pemetrexed, raltitrexed and pralatrexate, pyrimidine analogues such as fluoruracil, gemcitabine, floxuridine, 5-fluorouracil and tegafur-uracil, and purine analogues, selective estrogen receptor modulators and estrogen receptor downregulators.
  • Furthermore, also therapeutic antibodies can be used as further combination partner. It may be any antibody that is useful in cancer therapy which is different from the anti-MUC1 antibody. In particular, the further antibody is approved for cancer treatment by an administration such as the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA, formerly EMEA) and the Bundesinstitut for Arzneimittel und Medizinprodukte (BfArM). Examples of the further antibody that can be used for combination treatment are anti-EGFR antibodies such as Cetuximab, Tomuzotuximab, Panitumumab, Zalutumumab, Nimotuzumab, Matuzumab and Necitumumab; anti-HER2 antibodies such as Trastuzumab, Timigutuzumab and Pertuzumab; anti-VEGF antibodies such as bevacizumab (Avastin); anti-CD52 antibodies such as alemtuzumab (Campath); anti-CD30 antibodies such as brentuximab (Adcetris); anti-CD33 antibodies such as gemtuzumab (Mylotarg); and anti-CD20 antibodies such as rituximab (Rituxan, Mabthera), tositumomab (Bexxar) and ibritumomab (Zevalin). Further exemplary antibodies suitable for combination with the cancer therapy described herein include antibodies against antigens selected from the group consisting of Thomsen-Friedenreich antigen (TFα, TFP), Tn, Lewis Y, CD44, folate receptor a, NeuGc-GM3 ganglioside, DLL-3, RANKL, PTK7, Notch-3, Ephrin A4, insulin-like growth factor receptor 1, activin receptor-like kinase-1, claudin-6, disialoganglioside GD2, endoglin, transmembrane glycoprotein NMB, CD56, tumor-associated calcium signal transducer 2, tissue factor, ectonucleotide pyrophosphatase/phosphodiesterase 3, CD70, P-cadherin, mesothelin, six transmembrane epithelial antigen of the prostate 1 (STEAP1), carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), nectin 4, guanylyl cyclase C, solute carrier family 44 member 4 (SLC44A4), prostate-specific membrane antigen (PSMA), zinc transporter ZIP6 (LIV1 (ZIP6)), SLIT and NTRK-like protein 6 (SLITRK6), trophoblast glycoprotein (TPBG; 5T4), Fyn3, carbonic anhydrase 9, NaPi2b, fibronectin extra-domain B, endothelin receptor ETB, VEGFR2 (CD309), tenascin c, collagen IV and periostin.
  • The anti-MUC1 antibody can further be combined with checkpoint antibodies, i.e. antibodies blocking or activating immunomodulatory targets. Thereby, inhibitory signals for an immune response can be blocked and/or activating signals can be triggered. Examples of respective targets include CD40, CD3, CD137 (4-1BB), OX40, GITR, CD27, CD278 (ICOS), CD154 (CD40 ligand), CD270 (HVEM) and CD258 (LIGHT) as activating targets, CTLA4, PD1, CD80, CD244, A2AR, B7-H3 (CD276), B7-H4 (VTCN1), BTLA, IDO, KIR, LAG3, TIM-3, VISTA and phosphatidylserine as inhibitory targets, and their respective ligands such as PDL1.
  • In further embodiments, the anti-MUC1 antibody can be combined with the treatment with immunomodulatory compounds such as chemokines, cytokines, growth factors and vaccines. Suitable cytokines in this respect include interferons such as interferon-α, interferon-β and interferon-γ, and interleukins. Suitable growth factors include G-CSF and GM-CSF.
  • The anti-MUC1 antibody preferably is used for treatment of a primary tumor, a recurrent tumor and/or metastases of such tumors, and in particular is used for treatment before, during or after surgery and for the prevention or treatment of metastases. The anti-MUC1 antibody in particular is for the treatment of a patient as adjuvant therapy. In certain embodiments, the anti-MUC1 antibody is for the treatment of a patient as neoadjuvant therapy or in a combined neoadjuvant-adjuvant therapy. Furthermore, the anti-MUC1 antibody is for the treatment of a patient as palliative therapy.
  • The cancer therapy with the anti-MUC1 antibody preferably results in inhibition of tumor growth and in particular reduction of tumor size. Furthermore, the occurrence of further metastases is prevented and/or their number is reduced by the treatment. The treatment preferably results in an increase in progression-free survival; and/or an increase in lifespan and thus the overall survival.
  • The present invention further provides methods of therapy, diagnosis, prognosis, detecting and/or monitoring of a disease using the antibody according to the invention. The embodiments and examples of the use of the antibody in medicine also apply likewise to the medical methods. In particular, a method for treating a disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the antibody according to the present invention is provided.
  • For example, the invention provides a method for treating cancer in a subject in need thereof comprising, administering to the subject with cancer a therapeutically effective amount of the antibody according to the invention. In specific embodiments, the cancer is characterized by expressing TA-MUC1. The cancer may be selected from the group consisting of ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemia, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
  • Furthermore, the invention provides a method for diagnosis, detecting or monitoring cancer, comprising the step of contacting a test sample with an antibody according to the invention.
  • Methods of Increasing the MUC1 Binding Affinity
  • In a further aspect, the invention provides a method of increasing the MUC1 binding affinity of an antibody comprising
      • (i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6,
  • the method comprising the step of substituting the amino acid residue at position 8 of CDR-H2 with any amino acid residue except asparagine, resulting in CDR-H2 having the amino acid sequence of SEQ ID NO: 2.
  • The antibody which MUC1 binding affinity is to be increased in particular is an antibody capable of binding to MUC1 as described herein, except that it comprises an asparagine at position 8 of the CDR-H2 sequence.
  • In certain embodiments, the heavy chain variable region of the antibody which MUC1 binding affinity is to be increased comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 11. Especially, the heavy chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 11. In these embodiments, the heavy chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 1, 8 and 3. Hence, any sequence deviations to SEQ ID NO: 11 are located in the framework regions, but not in the CDRs. In particular, the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 11.
  • In certain embodiments, the light chain variable region of the antibody which MUC1 binding affinity is to be increased comprises an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12. Especially, the light chain variable region comprises an amino acid sequence which is at least 95%, in particular at least 98% identical to the amino acid sequence of SEQ ID NO: 12. In these embodiments, the light chain variable region still comprises CDRs having the amino acid sequences of SEQ ID NOs: 4, 5 and 6. Hence, any sequence deviations to SEQ ID NO: 12 are located in the framework regions, but not in the CDRs. In particular, the light chain variable region comprises the amino acid sequence of SEQ ID NO: 12.
  • In specific embodiments, the heavy chain variable region of the antibody which MUC1 binding affinity is to be increased has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 11, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 8 and 3, and the light chain variable region has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6. In particular, the heavy chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 11, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 1, 8 and 3, and the light chain variable region has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, wherein the CDRs still have the amino acid sequences of SEQ ID NOs: 4, 5 and 6.
  • For example, the antibody which MUC1 binding affinity is to be increased is an anti-MUC1 antibody as disclosed in WO 2004/065423 A2 or WO 2011/012309 A1. In particular, the antibody which MUC1 binding affinity is to be increased is gatipotuzumab or PankoMab.
  • The antibody which MUC1 binding affinity is increased in particular is an antibody capable of binding to MUC1 as described herein.
  • In certain embodiments, MUC1 binding is as described herein. Increasing the MUC1 or TA-MUC1 binding affinity in particular refers to an increase of at least 10%, at least 20%, at least 33% or at least 50%. In preferred embodiments, MUC1 binding affinity is increased by at least 50%. The MUC1 binding affinity may be determined as described in the examples, especially using surface plasmon resonance analysis or switchSENSE® Technology (DRX2 Biosensor, manufactured by Dynamic Biosensors GmbH), as described, e.g., in example 4a and b.
  • In certain embodiments, the step of substituting the amino acid residue at position 8 of CDR-H2 is achieved by introducing a mutation into the nucleic acid coding for the antibody, wherein the mutation is introduced in the codon coding for said amino acid residue. Introducing the mutation can be done by any method. Several suitable methods are known in the art and the skilled person is capable of performing the necessary tasks to introduce the mutation. The antibody with increased MUC1 binding affinity can then be obtained by expressing the mutated nucleic acid, for example in a host cell. Nucleic acids, host cells and methods for producing the antibody are described herein and can be used for the method for increasing the MUC1 binding affinity.
  • In specific embodiments, the method of increasing the MUC1 binding affinity of an antibody comprises the steps of
      • (a) providing a nucleic acid coding for the antibody which MUC1 binding affinity is to be increased
      • (b) introducing a mutation into said nucleic acid to produce a mutated nucleic acid, wherein the mutation is introduced in the codon coding for the amino acid residue at position 8 of CDR-H2 so that said codon codes for any amino acid residue except asparagine; and
      • (c) expressing the mutated nucleic acid to produce an antibody with increased MUC1 binding affinity.
  • The present invention further provides a method of producing an antibody with increased MUC1 binding affinity, comprising
      • (a) providing a nucleic acid coding for an antibody which comprises
        • (i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
        • (ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6;
      • (b) introducing a mutation into said nucleic acid to produce a mutated nucleic acid, wherein the mutation is introduced in the codon coding for the amino acid residue at position 8 of CDR-H2 so that said codon codes for any amino acid residue except asparagine; and
      • (c) producing the antibody with increased MUC1 binding affinity by expressing the mutated nucleic acid in a host cell.
  • The embodiments, features and examples described herein for the other aspects, especially for the method of increasing the MUC1 binding affinity of an antibody, also likewise apply to the method of producing an antibody with increased MUC1 binding affinity.
  • In certain embodiments, the method of producing an antibody with increased MUC1 binding affinity further comprises a step (d) of processing the antibody with increased MUC1 binding affinity.
  • For example, processing the antibody with increased MUC1 binding affinity may include isolating the antibody from the cell culture. Isolation of the antibody in particular refers to the separation of the antibody from the remaining components of the cell culture. Separation of the antibody from the cell culture medium may be performed, for example, by chromatographic methods. Suitable methods and means for isolating antibodies are known in the art and can be readily applied by the skilled person.
  • The obtained antibody may optionally be subject to further processing steps such as e.g. modification steps such as chemical or enzymatic coupling of a further agent to the antibody, and/or formulation steps in order to produce the antibody in the desired quality and composition. Such further processing steps and methods are generally known in the art.
  • In further embodiments, step (d) additionally comprises the step of providing a pharmaceutical formulation comprising the antibody. Providing a pharmaceutical formulation comprising the antibody or formulating the antibody as a pharmaceutical composition in particular comprises exchanging the buffer solution or buffer solution components of the composition comprising the antibody. Furthermore, this step may include lyophilization of the antibody. In particular, the antibody is transferred into a composition only comprising pharmaceutically acceptable ingredients.
  • Specific Embodiments
  • In the following, specific embodiments of the present invention are described.
  • Embodiment 1. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • Embodiment 2. The antibody according to Embodiment 1, wherein the amino acid at position 8 of the CDR-H2 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine, especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine, in particular glutamine.
  • Embodiment 3. The antibody according to Embodiment 1, wherein the amino acid at position 8 of the CDR-H2 is glutamine, histidine, arginine, tryptophan, or lysine.
  • Embodiment 4. The antibody according to Embodiments 1 to 3, wherein the CDR-H2 has the amino acid sequence of SEQ ID NO: 7.
  • Embodiment 6. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which
        • (a) has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 9, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region, which
        • (a) has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • Embodiment 7. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which
        • (a) has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 9, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region, which
        • (a) has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • Embodiment 8. The antibody according to Embodiment 6 or 7, wherein the amino acid at position 8 of the CDR-H2 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine, especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine, in particular glutamine.
  • Embodiment 9. The antibody according to Embodiment 7 or 8, wherein the amino acid at position 8 of the CDR-H2 is glutamine, histidine, arginine, tryptophan, or lysine.
  • Embodiment 10. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which
        • (a) has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 10, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 7 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region, which
        • (a) has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • Embodiment 11. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which
        • (a) has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 10, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 7 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region, which
        • (a) has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • Embodiment 12. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which has the amino acid sequence of SEQ ID NO: 9, and
      • (ii) a light chain variable region, which has the amino acid sequence of SEQ ID NO: 12.
  • Embodiment 13. The antibody according to Embodiment 12, wherein the amino acid at position 57 of SEQ ID NO: 9 is selected from the group consisting of glutamine, alanine, valine, histidine, tryptophan, tyrosine, lysine and arginine, especially glutamine, histidine, tryptophan, tyrosine, lysine and arginine, in particular glutamine.
  • Embodiment 14. The antibody according to Embodiment 12, wherein the amino acid at position 57 of SEQ ID NO: 9 is glutamine, histidine, arginine, tryptophan, or lysine.
  • Embodiment 15. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which has the amino acid sequence of SEQ ID NO: 10, and
      • (ii) a light chain variable region, which has the amino acid sequence of SEQ ID NO: 12.
  • Embodiment 16. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which
        • (a) has an amino acid sequence which is at least 90% or at least 95% identical to the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 or 7 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region, which
        • (a) has an amino acid sequence which is at least 90% or at least 95% identical to the amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • Embodiment 17. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which has the amino acid sequence represented by amino acid Nos 20 to 136 of SEQ ID NO: 20 or 23, and
      • (ii) a light chain variable region, which has amino acid sequence represented by amino acid Nos 21 to 133 of SEQ ID NO: 21.
  • Embodiment 18. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain, which
        • (a) has an amino acid sequence which is at least 90% or at least 95% identical to the amino acid sequence of SEQ ID NO: 15, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 or 7 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region, which
        • (a) has an amino acid sequence which is at least 90% or 95% identical to the amino acid sequence of SEQ ID NO: 16, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • Embodiment 19. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which has the amino acid sequence of SEQ ID NO: 15 or SEQ ID NO: 22, and
      • (ii) a light chain variable region, which has the amino acid sequence of SEQ ID NO: 16.
  • Embodiment 20. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain variable region, which
        • (a) has an amino acid sequence which is at least 90% or at least 95% identical to the amino acid sequence represented by amino acid Nos 20 to 460 of SEQ ID NO: 20, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 or 7 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region, which
        • (a) has an amino acid sequence which is at least 90% or 95% identical to the amino acid sequence represented by amino acid Nos 21 to 239 of SEQ ID NO: 21, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
  • Embodiment 21. An antibody capable of binding to MUC1, which comprises
      • (i) a heavy chain, which has the amino acid sequence represented by amino acid Nos 20 to 460 of SEQ ID NO: 20 or 23, and
      • (ii) a light chain, which has amino acid sequence represented by amino acid Nos 21 to 239 of SEQ ID NO: 21.
  • Embodiment 22. The antibody according to any one of Embodiments 1 to 21, wherein the antibody comprises at least one heavy chain, comprising the heavy chain variable region, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain.
  • Embodiment 23. The antibody according to any one of Embodiments 1 to 21, wherein the antibody comprises two heavy chains, each comprising the heavy chain variable region, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain.
  • Embodiment 24. The antibody according to Embodiment 22 or 23, wherein the antibody is an IgG-type antibody, in particular an IgG1, IgG2 or IgG4-type antibody.
  • Embodiment 25. The antibody according to any one of Embodiments 1 to 24, wherein the antibody comprises at least one light chain, comprising the light chain variable region and a CL domain.
  • Embodiment 26. The antibody according to any one of Embodiments 1 to 24, wherein the antibody comprises two light chains, each comprising the light chain variable region and a CL domain.
  • Embodiment 274. The antibody according to Embodiment 25 or 26, wherein the light chain is a κ-type light chain.
  • Embodiment 28. The antibody according to any one of Embodiments 1 to 27, wherein the antibody does not comprise an N-glycosylation site in the CH2 domain.
  • Embodiment 29. The antibody according to any one of Embodiments 1 to 27, wherein the antibody comprises an N-glycosylation site in the CH2 domain of the antibody heavy chains.
  • Embodiment 30. The antibody according to Embodiment 29, wherein the antibody has a glycosylation pattern having one or more of the following characteristics
      • (i) a relative amount of glycans carrying a bisecting GlcNAc residue of at least 0.5% of the total amount of glycans attached to the glycosylation sites of the antibody in a composition;
      • (ii) a relative amount of glycans carrying at least one galactose residue of at least 30% of the total amount of glycans attached to the glycosylation sites of the antibody in a composition;
      • (iii) a relative amount of glycans carrying a core fucose residue of at least 60% of the total amount of glycans attached to the glycosylation sites the antibody in a composition.
  • Embodiment 31. The antibody according to Embodiment 29, wherein the antibody has a glycosylation pattern having one or more of the following characteristics
      • (i) a relative amount of glycans carrying a bisecting GlcNAc residue of at least 0.5% of the total amount of glycans attached to the glycosylation sites of the antibody in a composition;
      • (ii) a relative amount of glycans carrying at least one galactose residue of at least 30% of the total amount of glycans attached to the glycosylation sites of the antibody in a composition;
      • (iii) a relative amount of glycans carrying a core fucose residue of 40% or less of the total amount of glycans attached to the glycosylation sites of the antibody in a composition.
  • Embodiment 32. The antibody according to any one of Embodiments 1 to 31, comprising a further agent conjugated thereto.
  • Embodiment 33. The antibody according to Embodiment 32, wherein the further agent is a chemotherapeutic agent which is coupled to the antibody.
  • Embodiment 34. The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of a maytansinoid, a DNA damaging agent, a DNA alkylating agent and a DNA minor groove binder.
  • Embodiment 35. The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of maytansinol, N2′-deacetyl-N2′-(3-mercapto-1-oxopropyl)-maytansine (DM1), N2′-deacetyl-N2′-(4-mercapto-1-oxopentyl)-maytansine (DM3), and N2′-deacetyl-N2′-(4-methyl-4-mercapto-1-oxopentyl)-maytansine (DM4).
  • Embodiment 36. The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of monomethyl auristatin F (MMAF), monomethyl auristatin E (MMAE) and auristatin T.
  • Embodiment 37. The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of pyrrolobenzodiazepine (PBD), pyrrolobenzodiazepine dimer (PBD dimer), duocarmycin, duocarmycin-hydroxybenzamide-azaindole (DUBA), seco-duocarmycin-hydroxybenzamide-azaindole (seco-DUBA) and doxorubicin.
  • Embodiment 38. The antibody according to Embodiment 33, wherein the chemotherapeutic agent is selected from the group consisting of indolinobenzodiazepine and oxazolidinobenzodiazepine.
  • Embodiment 39. The antibody according to Embodiment 33, wherein the chemotherapeutic agent is calicheamicin.
  • Embodiment 40. The antibody according to Embodiment 32, wherein the further agent is a polypeptide or protein which is fused to a polypeptide chain of the antibody.
  • Embodiment 41. The antibody according to Embodiment 40, wherein the antibody comprises two antibody heavy chains and two antibody light chains and a further agent being a polypeptide or protein is fused to each of the C termini of said antibody heavy chains or to each of the C termini of said antibody light chains.
  • Embodiment 42. The antibody according to Embodiment 40 or 41, wherein the further agent is selected from the group consisting of cytokines, chemokines, other antibodies, antigen binding fragments, enzymes and binding domains.
  • Embodiment 43. The antibody according to Embodiment 41, wherein the further agent is a scFv fragment specifically binding to CD3, and one of said further agent is fused to the C terminus of each antibody heavy chain.
  • Embodiment 44. The antibody according to Embodiment 41, wherein the further agent is a scFv fragment specifically binding to PDL1, and one of said further agent is fused to the C terminus of each antibody light chain.
  • Embodiment 45. A nucleic acid encoding the antibody according to any one of Embodiments 1 to 44.
  • Embodiment 46. An expression cassette or vector comprising the nucleic acid according to Embodiment 75 and a promoter operatively connected with said nucleic acid.
  • Embodiment 47. A host cell comprising the nucleic acid according to Embodiment 45 or the expression cassette or vector according to Embodiment 46.
  • Embodiment 48. A pharmaceutical composition comprising the antibody or conjugate according to any one of Embodiments 1 to 44 and one or more further components selected from the group consisting of solvents, diluents, and excipients.
  • Embodiment 49. The antibody according to any one of Embodiments 1 to 44 or the pharmaceutical composition according to Embodiment 48 for use in medicine.
  • Embodiment 50. The antibody according to any one of Embodiments 1 to 44 or the pharmaceutical composition according to Embodiment 48 for use in the treatment, prognosis, diagnosis, detecting and/or monitoring of diseases associated with abnormal cell growth such as cancer; infections such as bacterial, viral, fungal or parasitic infections; inflammatory diseases such as autoimmune diseases and inflammatory bowel diseases; and diseases associated with a reduce immune activity such as immunodeficiencies.
  • Embodiment 51. The antibody or pharmaceutical composition according to Embodiment 50 for use in the treatment of cancer, in particular the cancer expressing TA-MUC1, wherein the cancer is selected from the group consisting of cancer of the ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemia, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
  • Embodiment 52. The antibody or pharmaceutical composition according to Embodiment 50 for use in the treatment of infections, wherein the infection is selected from the group consisting of bacterial infections, viral infections, fungal infections and parasitic infections.
  • Embodiment 53. The antibody or pharmaceutical composition according to Embodiment 50 for use in the treatment of autoimmune diseases, wherein the autoimmune disease is selected from the group consisting of celiac disease, diabetes mellitus type 1, Graves disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis and systemic lupus erythematosus.
  • Embodiment 54. A method of increasing the MUC1 binding affinity of an antibody comprising
      • (i) a heavy chain variable region, which
        • (a) has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 11, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region, which
        • (a) has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6;
  • the method comprising the step of substituting the amino acid residue at position 8 of CDR-H2 with any amino acid residue except asparagine, resulting in CDR-H2 having the amino acid sequence of SEQ ID NO: 2.
  • Embodiment 55. A method of increasing the MUC1 binding affinity of an antibody comprising
      • (i) a heavy chain variable region, which
        • (a) has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 11, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
      • (ii) a light chain variable region, which
        • (a) has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, and
        • (b) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6;
  • the method comprising the step of substituting the amino acid residue at position 8 of CDR-H2 with any amino acid residue except asparagine, resulting in CDR-H2 having the amino acid sequence of SEQ ID NO: 2.
  • Embodiment 56. The method according to embodiment 54 or 55, wherein substituting the amino acid residue at position 8 of CDR-H2 is achieved by introducing a mutation into the nucleic acid coding for the antibody, wherein the mutation is introduced in the codon coding for said amino acid residue.
  • Embodiment 57. The method according to any one of Embodiments 54 to 56, comprising the steps of
      • (a) providing a nucleic acid coding for the antibody which MUC1 binding affinity is to be increased;
      • (b) introducing a mutation into said nucleic acid to produce a mutated nucleic acid, wherein the mutation is introduced in the codon coding for the amino acid residue at position 8 of CDR-H2 so that said codon codes for any amino acid residue except asparagine; and
      • (c) expressing the mutated nucleic acid to produce an antibody with increased MUC1 binding affinity.
  • Embodiment 58. A method of producing an antibody with increased MUC1 binding affinity, comprising
      • (a) providing a nucleic acid coding for an antibody which comprises
        • (i) a heavy chain variable region, which
          • (i1) has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 11, and
          • (i2) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
        • (ii) a light chain variable region, which
          • (ii1) has an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12, and
          • (ii2) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6;
      • (b) introducing a mutation into said nucleic acid to produce a mutated nucleic acid, wherein the mutation is introduced in the codon coding for the amino acid residue at position 8 of CDR-H2 so that said codon codes for any amino acid residue except asparagine; and
      • (c) producing the antibody with increased MUC1 binding affinity by expressing the mutated nucleic acid in a host cell.
  • Embodiment 59. A method of producing an antibody with increased MUC1 binding affinity, comprising
      • (a) providing a nucleic acid coding for an antibody which comprises
        • (i) a heavy chain variable region, which
          • (i1) has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 11, and
          • (i2) comprises the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
        • (ii) a light chain variable region, which
          • (ii1) has an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO: 12, and
          • (ii2) comprises the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6;
      • (b) introducing a mutation into said nucleic acid to produce a mutated nucleic acid, wherein the mutation is introduced in the codon coding for the amino acid residue at position 8 of CDR-H2 so that said codon codes for any amino acid residue except asparagine; and
      • (c) producing the antibody with increased MUC1 binding affinity by expressing the mutated nucleic acid in a host cell.
  • Embodiment 60. The method according to any one of Embodiments 54 to 59, wherein the antibody comprises two heavy chains, each comprising the heavy chain variable region, a CH1 domain, a hinge region, a CH2 domain and a CH3 domain.
  • Embodiment 61. The method according to Embodiment 60, wherein the antibody is an IgG-type antibody, in particular an IgG1, IgG2 or IgG4-type antibody.
  • Embodiment 62. The method according to any one of Embodiments 54 to 60, wherein the antibody comprises two light chains, each comprising the light chain variable region and a CL domain.
  • Embodiment 63. The method according to Embodiment 62, wherein the light chain is a κ-type light chain.
  • Embodiment 64. The method according to any one of Embodiments 54 to 63, wherein the antibody with increased MUC1 binding affinity is an antibody as defined in any one of Embodiments 1 to 44.
  • Embodiment 65. A method for treating cancer in a subject in need thereof comprising, administering to the subject with cancer, in particular cancer expressing TA-MUC1, a therapeutically effective amount of the antibody according to any one of Embodiments 1 to 44 or the composition according to Embodiment 48.
  • Embodiment 66. The method for treating cancer according to Embodiment 65, wherein the cancer is selected from the group consisting of ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemia, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
  • FIGURES
  • FIG. 1 shows ELISA binding curves of the anti-MUC1 antibodies to different MUC1 peptides. (A) shows antigen binding of PankoMab N54Q (PM-N54Q) lacking Fab glycosylation and PankoMab comprising Fab glycosylation (PM) to the MUC1 peptide comprising the epitope sequence PDTR. The threonine of the MUC1 peptide is glycosylated with Tn, sTn, TF or sTF. (B) shows binding of PankoMab and PankoMab N54Q to the MUC1 peptide comprising the epitope sequence variant PESR. The serine of the MUC1 peptide is glycosylated with Tn. (C) shows binding of PankoMab N54Q to the MUC1 peptide comprising the epitope sequence PDTR. The threonine of the MUC1 peptide is glycosylated with Tn or not glycosylated. (D) shows binding of several N54X variants to Tn-PDTR MUC1 peptide compared to PankoMab comprising Fab glycosylation diluted from cell culture supernatant of transiently transfected cells. (E) shows binding curves of three purified N54X variants without Fab glycosylation in comparison to PankoMab with Fab glycosylation on Tn-PDTR, TF-PDTR and non-glycosylated PDTR MUC1 peptide. (F) shows binding of two framework variants of PM-N54Q to Tn-PDTR MUC1 peptide compared to PankoMab with Fab glycosylation. For framework variant mf-a nine amino acids are mutated in the VH and three in the VL framework, for mf-b also nine amino acids are mutated in the VH and four in the VL framework.
  • FIG. 2 shows surface plasmon resonance (Biacore) binding of the anti-MUC1 antibodies PM and PM-N54Q to a Tn-glycosylated PDTR-MUC1 peptide. The maximal binding signal of different concentrations of PankoMab N54Q and PankoMab are plotted against the antibody concentration.
  • FIG. 3 shows results of Fluorescence Proximity Sensing on DRX instrument. Association and dissociation curves are shown. (A) PM with Fab glycosylation compared to (B) PM-N54Q without Fab glycosylation.
  • FIG. 4 shows an SDS acrylamide gel of an electrophoretic separation of PankoMab N54Q and PankoMab under non-reducing (left) and reducing (right) conditions. Lane 1: PankoMab N54Q after capture step; lane 2: PankoMab N54Q after polishing step; lane 3: PankoMab after capture step; lane 4: PankoMab after polishing step; lane 5: molecular weight marker.
  • FIG. 5 shows the Coomassie blue stained gel of an isoelectric focusing assay with PankoMab N54Q lacking Fab glycosylation and PankoMab being Fab-glycosylated. Lane 1: PankoMab with Fab glycosylation; lane 2: PankoMab N54Q without Fab glycosylation.
  • FIG. 6 shows anti-MUC1 antibody binding to Fcγ receptor IIIa. Increasing concentrations of the antibody PankoMab N54Q or PankoMab displace rabbit-anti-mouse coupled acceptor beads from FcγRIIIa loaded donor beads, thereby reducing the detected chemiluminescence. In FIG. 6A low-fucosylated antibodies and in FIG. 6B high-fucosylated antibodies were applied into the assay.
  • FIG. 7 shows binding of the anti-MUC1 antibodies PM-N54Q, PM-N54D and PM with Fab glycosylation to the tumor cell lines (A) CaOV-3 and (B) HSC-4 as analyzed by flow cytometry.
  • FIG. 8 shows the amino acid sequence of the heavy chain of the humanized antibody PankoMab N54Q (SEQ ID No: 15, wherein the amino acid at position 57 is Gln, namely SEQ ID No: 22).
  • FIG. 9 shows the amino acid sequence of the light chain of the humanized antibody PankoMab N54Q (SEQ ID No: 16).
  • FIG. 10 shows the amino acid sequence of the heavy chain of the humanized antibody PankoMab (SEQ ID No: 19).
  • FIG. 11 shows the amino acid sequence of the heavy chain of chimeric antibody PankoMab N54Q (SEQ ID No: 20, wherein the amino acid at position 76 is Gln, namely SEQ ID No: 23).
  • FIG. 12 shows the amino acid sequence of the light chain of chimeric antibody PankoMab N54Q (SEQ ID No: 21).
  • EXAMPLES Example 1: Production of Anti-MUC1 Antibodies
  • The nucleic acid sequence of the heavy chain of humanized PankoMab antibody (see, e.g., WO 2011/012309) was modified by mutating the codon for Asn54 according to the Kabat/EU numbering system (amino acid position 57 in SEQ ID NO: 11) into the codon for any amino acid except Asn, especially for Gln.
  • 1) Production of the Anti-MUC1 Antibodies in a Human Myeloid Leukemia Derived Cell Line
  • Vectors comprising the coding sequences of the γ1-type heavy chain and the κ-type light chain of the mutated antibodies were transfected into the human myeloid leukemia derived cell line NM-H9D8 (DSM ACC2806). The different αMUC1-antibodies comprising the N54X mutation (PankoMab N54X/PM-N54X whereby X is any amino acid except N/Asn) or amino acid mutations in the framework sequences of the VH and VL were expressed in the obtained clones, producing the constructs with a human glycosylation pattern. The concentration of the αMUC1-antibodies in the supernatant was determined by Octet measurement using Protein A coated pins or were quantified by UV280 absorbance after purification by protein A chromatography. The binding characteristics of the different αMUC1-antibodies were determined by Antigen-ELISA (see example 2), and selected purified antibodies were also analyzed by Scatchard analysis (see example 3), by Biacore (see example 4a), by DRX2 switchSENSE® technology (see example 4b), or by flow cytometry (example 7).
  • In addition, PM-N54Q and non-mutated PankoMab with Fab-glycosylation were also expressed in the human myeloid leukemia derived cell line NM-H9D8-E6Q12 (DSM ACC2856) expressing antibody with reduced fucose. Together with the same antibodies expressed in NM-H9D8, these antibodies were purified and analyzed in example 6 for their binding behavior to Fc gamma receptor Ill A.
  • 2) Production of the Anti-MUC1 Antibody in CHO Cell Line
  • PM-N54Q encoding sequences (nucleotide sequence of heavy chain of PM-N54Q represented by SEQ ID NO: 17 and nucleotide sequence of light chain of PM-N54Q represented by SEQ ID NO: 18) which was synthesized by GeneArt™ of ThermoFisher scientific were cloned into expression vectors and resulting plasmids were electro-transfected into CHO cells. Pooled cells grown under selection pressure were applied to manufacture PM-N54Q mutant antibody with general procedures.
  • Example 2: Antigen ELISA
  • The antigen binding characteristics of PankoMab N54X, wherein the N-glycosylation site in the Fab part is knocked out, was compared to PankoMab having an N-glycosylation site in its Fab part.
  • Binding characteristics of the Fab-deglycosylated version of the MUC1-specific antibody PankoMab (PM-N54Q) compared to the (glycosylated) PankoMab-GEX® were analyzed using differently glycosylated and the non-glycosylated MUC1-derived tandem repeat peptides in ELISA studies. In principle, both antibodies show the same gradation by means of binding to glycosylated PDTR peptides (APPAHGVTSAPD-T(X)-RPAPGSTAPPAHGVTSA) with different glycosylations at T: Strongest binding was observed to the PDTR peptide carrying a Galβ1-3GalNAcalpha (TF) followed by sialylated TF GalNAcalpha (Tn) O-glycosylation. Binding to sialylated GalNAcalpha(sTn)O-glycosylation was significantly lower. As PankoMab-GEX®, PM-N54Q showed only little binding activity to non-glycosylated MUC1 PDTR peptide indicating adequate tumor specificity (FIG. 1C).
  • However, in comparison to PankoMab-GEX® four-fold higher binding was found for PM-N54Q in the TA-MUC1 antigen ELISA using the biotinylated glycopeptide carrying a GalNAcalpha (Tn)O-glycan. PM-N54Q binds about seven-fold better to the same MUC1 peptide when glycosylated with sialylated GalNAcalpha (sTn). The binding to Galβ1-3GalNAcalpha (TF) and sialylated TF (sTF) at the threonine of the PDTR-sequence (FIG. 1A) was two-fold better for PM-N54Q.
  • Both antibodies show strongly diminished binding to the MUC1 peptide variant APPAHGVTSAPE-S(Tn)-RPAPGSTAPPAHGVTSA with Tn glycosylation at the serine compared to that at PDT(Tn)R-peptide. However, also here the Fab-deglycosylated PM-N54Q binds significantly stronger than PankoMab-GEX® (FIG. 1B).
  • Different other Fab-deglycosylated PM-N54X variants were compared to PankoMab having an N-glycosylation in its Fab part. First, all variants were compared directly from the supernatant, without purification. The concentration was determined by Octet. All PM-N54X variants bound better than Fab-glycosylated PM. In addition, a clear trend depending on the chemical properties of the amino acid side chain was visible. Carboxylic acid groups at the side chain showed the lowest binding enhancement. Best binding was observed for amino acids with one or two nitrogens (as primary or secondary amines) (FIG. 1D).
  • In addition, selected Fab-deglycosylated variants (PM-N54H, -W and -Q) were purified by Protein A chromatography and analyzed on ELISA (FIG. 1E). The improvement of binding to TF-MUC1 peptide is about 5- to 8-fold and to Tn-MUC1 peptide about 2- to 3-fold compared to PankoMab with Fab-glycosylation, respectively.
  • Furthermore, two different framework variants of the PM-N54Q were analyzed for the binding to the Tn-glycosylated PDTR-MUC1 peptide in ELISA (see FIG. 1F). The framework variant mf-a carries nine amino acid mutations in the VH and three in the VL framework; the variant mf-b carries also nine amino acid mutations in the VH and four in the VL framework. Both mutated variants show similar binding compared to the PM-N54Q antibody.
  • Example 3: Saturation Binding Analyses of Anti-MUC1 Antibodies to MCF-7 and ZR-75-1 Cells
  • Two factors are especially critical for the therapeutic suitability of an antibody: the affinity and number of binding sites of an antibody on tumor cells.
  • Binding of the Fab-deglycosylated version of the MUC1-specific antibody PankoMab (PM-N54Q) on TA-MUC-1 positive human tumor cell lines was evaluated using radiolabeled antibodies by saturated binding analysis on the human mamma carcinoma cell lines ZR-75-1 and MCF-7 in comparison to Fab-glycosylated PankoMab-GEX®.
  • The antibodies were chelated with a 12-fold molar excess of p-SCN-Benzyl-DTPA in 50 mM sodium carbonate, 150 mM NaCl, pH 8.7, for 2 h at 37° C., followed by over-night incubation at 2-8° C. Free chelator was removed over desalting columns and dead-end filtration (50 kDa cut-off, 6× buffer exchange to PBS). The chelated antibodies were radiolabeled with carrier-free 111In (2 μCi/μg antibody) for 1 h at 37° C. in 6 mM phosphate, 1.6 mM KCl, 80 mM NaCl, 0.2 M Na-acetate, 0.1 M HCl. The preparations were neutralized by addition of 8-9fold volume of 10× concentrated PBS. About 1/50 volume of fetal bovine serum were added to the neutralized labelled antibody preparation. Per cell binding approach 1*106 cells were used. Several concentrations of labelled antibodies were added to the pelleted cells (30-1000 ng/200 μL in 1% BSA/PBS). The resuspended cell-antibody mixtures were measured in a gamma-counter and incubated 1 h at 4° C. Cells with bound antibodies were separated by centrifugation and washed with 1% BSA/PBS for another hour at 4° C. The cell pellet was then measured for bound 111In-labelled antibody in a gamma counter. Evaluation was performed by “one-site specific ka” in GraphPad Prism. The obtained data are summarized in Table 1. The data show the high affinity and very high number of binding sites of PM-N54Q on these tumor cells. The binding was more than 2.5-fold higher than for PankoMab-GEX® and also the number of binding sites was slightly increased.
  • TABLE 1
    Association constant and antigen
    binding sites on MUC1+ tumor cells
    ZR-75-1 MCF-7
    Kass [1/M]
    PM w Fab glyc. 1.2 × 107 3 × 107
    PM N54Q w/o Fab glyc. 3.4 × 107 7.8 × 107
    Binding sites
    PM w Fab glyc.  20 × 105 0.6 × 105
    PM N54Q w/o Fab glyc.  30 × 105 0.9 × 105
  • Example 4a: Surface Plasmon Resonance (BiaCore) Analysis
  • Binding of the Fab-deglycosylated version of the MUC1-specific antibody PankoMab (PM-N54Q) on TA-MUC-1 derived glycosylated peptide was evaluated by surface plasmon resonance analysis (Biacore). A streptavidin sensor chip was coated with biotinylated TA-MUC1 peptide (Tn glycosylated or not glycosylated). PankoMab and PM-N54Q were diluted sequentially 1:3 from 3,600 to 4.9 nM in HPS-EP. The dilutions were injected at 50 μL/min. Maximal binding of each concentration was determined as response units (RU), respectively, and evaluated with GraphPad Prism using “one-site specific binding”. FIG. 2 shows the obtained binding curves with PM-N54Q compared to PankoMab-GEX®. Affinities (K) of 388 nM and 652 nM were calculated for PM-N54Q and PankoMab-GEX®, respectively. Therefore, in this experimental setting a nearly two-fold increase in affinity was detectable.
  • Example 4b: Fluorescence Proximity Sensing (by DRX2, Dynamic Biosensors)
  • A new method to determine binding constants and affinity is the fluorescence proximity sensing using single stranded DNA (96mer) spotted on a chip and complementary DNA coupled to a ligand. In the present study streptavidin was used as a ligand to capture biotinylated TA-MUC1 peptides. Binding of PankoMab to the peptides resulted in a fluorescence change. On- and off-rates can be calculated during association and dissociation. Due to a higher sensitivity faster interactions can be monitored compared to surface plasmon resonance. This results in binding kinetics different from SPR but more comparable to the “gold standard” method KinExA, measured in a liquid system.
  • PankoMab and PM-N54Q were diluted from 300 nM in 1:9 steps to 3.67 nM in PE140 buffer and applied to the chip-bound peptides. Binding curves were evaluated by mono-exponential global fit (instrument software). Binding curves of PM and PM-N54Q are exemplarily shown in FIGS. 3A and B. Calculated affinities of PankoMab variants are shown in Table 2:
  • TABLE 2
    Dissociation constants of PankoMab variants to antigen peptide
    PankoMab variant KD
    PM with Fab glycosylation 4.1 nM
    PM-N54D 1.9 nM
    PM-N54Q 1.6 nM
    PM-N54H 0.6 nM
  • Example 5: Biochemical Characterization
  • Non-reducing and reducing SDS-PAGE is used to analyze purity and identity of an antibody. The band pattern in non-reducing gels shows the major band at about 160 kDa and methodical artefacts of heavy and light chains and combinations thereof (25, 50-55, 75, 110, 135 kDa). Reducing gels show distinct light and heavy chain bands at 25 and 50-55 kDa. Due to lack of the Fab glycosylation PM-N54Q has a smaller heavy chain, as expected (see FIG. 4, right).
  • The charge profile is clearly different, as shown by isoelectric focusing (IEF; see FIG. 5). The Fab glycosylation is considerably sialylated, whereas the Fc glycosylation is only minimally sialylated. Thus PankoMab-GEX® has more charged isoforms than PM-N54Q, reflecting its higher level of negatively charged sialic acids in the Fab part.
  • Example 6: Fcγ Receptor Binding
  • FcγR binding assays for FcγRIIIa (CD16a) are based on the AlphaScreen® technology of PerkinElmer. The AlphaScreen® platform relies on simple bead-based technology of PerkinElmer and is a more efficient alternative to traditional ELISA since no washing steps are necessary.
  • For the receptor binding assays, His-tagged FcγRIIIa (Glycotope GmbH) is captured by Ni-chelate donor beads. Anti-MUC1 antibodies and rabbit-anti-mouse coupled acceptor beads compete for binding to FcγR. In case of interaction of FcγR with rabbit-anti-mouse-bound acceptor beads, donor and acceptor beads come into close proximity which leads, upon laser excitation at 680 nm, to light emission. A maximum signal is achieved (signalmax) without a competitor. In case of competition, where a test antibody binds to FcγR, the signalmax is reduced in a concentration-dependent manner. Chemiluminescence was quantified by measurement at 520-620 nm (AlphaScreen® method) using an EnSpire 2300 multilabel reader (PerkinElmer). All results were expressed as the mean±standard deviation of duplicate samples. The data were evaluated and calculated using non-linear curve fitting (sigmoidal dose-response variable slope) with GraphPad Prism 5 software. As a result, a concentration dependent sigmoidal curve was obtained, which is defined by top-plateau, bottom-plateau, slope and EC50.
  • As shown in FIGS. 6A and B, the FcγRIIIa binding affinity was comparable for PankoMab N54Q and PankoMab whereby in Figure A low-fucosylated antibodies and in Figure B high-fucosylated antibodies were applied into the assay. Hence, removal of the Fab glycosylation did not affect receptor interaction of the antibody.
  • Example 7: Binding to Cellular TA-MUC1
  • N54Q and N54D were transiently expressed and purified by protein A chromatography. Binding of the two variants to cell surface TA-MUC1 was compared to PM with Fab glycosylation using two different carcinoma cell lines. The tongue squamous cell carcinoma line HSC-4 expresses TA-MUC1 to a medium degree and the ovarian carcinoma cell line CaOV-3 to a high degree. Tumor cells were incubated with antibodies in serial dilutions and bound antibodies were detected using a Phycoerythrin-conjugated goat anti-human IgG (heavy and light chain) antibody. A human IgG control was included to control for background staining. Binding was analyzed by flow cytometry.
  • The analyzed constructs PM, PM-N54Q and PM-N54D show strong and specific binding to the TA-MUC1 expressing HSC-4 and CaOV-3 cells compared to a human IgG1 control (FIG. 7). The binding of PM-N54D to the TA-MUC1high CaOV-3 cells was comparable to PM with Fab glycosylation while PM-N54Q showed a slightly better binding (FIG. 7A). Using HSC-4 carcinoma cells that express TA-MUC1 at an intermediate level, the variant PM-N54Q was clearly superior in binding to cellular TA-MUC1 compared to PM while PM-N54D showed an inferior binding compared to PM with Fab glycosylation (FIG. 7B).
  • Identification of the Deposited Biological Material
  • The cell lines DSM ACC 2806, DSM ACC 2807 and DSM ACC 2856 were deposited at the DSMZ—Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH, Inhoffenstralße 7B, 38124 Braunschweig (DE) by Glycotope GmbH, Robert-Rössle-Str. 10, 13125 Berlin (DE) on the dates indicated in the following table.
  • Name of the Accession Date of
    Cell Line Number Depositor Deposition
    NM-H9D8 DSM ACC 2806 Glycotope GmbH Sep. 15,
    2006
    NM-H9D8-E6 DSM ACC 2807 Glycotope GmbH Oct. 5, 2006
    NM-H9D8-E6Q12 DSM ACC 2856 Glycotope GmbH Aug. 8, 2007

Claims (40)

1. An antibody capable of binding to MUC1, which comprises
(i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
(ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
2. The antibody according to claim 1, wherein the amino acid at position 8 of SEQ ID NO: 2 is selected from the group consisting of glutamine, histidine, tryptophan, tyrosine, lysine and arginine, or wherein the CDR-H2 has the amino acid sequence of SEQ ID NO: 7
3. The antibody according to claim 1, wherein the heavy chain variable region has the amino acid sequence of SEQ ID NO: 9 or an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 9.
4. The antibody according to claim 1, wherein the heavy chain variable region has the amino acid sequence of SEQ ID NO: 10 or an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 10.
5. The antibody according to claim 1, wherein the light chain variable region has the amino acid sequence of SEQ ID NO: 12 or an amino acid sequence which is at least 90% identical to the amino acid sequence of SEQ ID NO: 12.
6. The antibody according to claim 1, wherein the heavy chain variable region of the antibody has the amino acid sequence of SEQ ID NO: 10 and the light chain variable region of the antibody has the amino acid sequence of SEQ ID NO: 12.
7. The antibody according to claim 1, wherein the antibody comprises an Fc region and preferably is an IgG1, IgG2 or IgG4-type antibody.
8. The antibody according to claim 1, wherein the antibody comprises a heavy chain having the amino acid sequence of SEQ ID NO: 15, in particular SEQ ID NO: 22, and a light chain having the amino acid sequence of SEQ ID NO: 16.
9. The antibody according to claim 7, wherein the antibody comprises a glycosylation pattern having one or more of the following characteristics:
(i) a detectable amount of glycans carrying a bisecting GlcNAc residue;
(ii) a relative amount of glycans carrying at least one galactose residue of at least 25% of the total amount of glycans attached to the Fc glycosylation sites of the antibody in a composition.
10. The antibody according to claim 1, wherein the antibody is obtainable by production in a mammalian cell.
11. The antibody according to claim 1, obtainable by production in a human cell line selected from the group consisting of NM-H9D8 (DSM ACC 2806), NM-H9D8-E6 (DSM ACC 2807), NM-H9D8-E6Q12 (DSM ACC 2856) and cell lines derived therefrom.
12. The antibody according to claim 1, wherein the antibody is obtainable by production in a CHO cell line or a cell line derived therefrom.
13. The antibody according to claim 1, wherein the antibody competes for the binding to TA-MUC1 with an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region having the amino acid sequence of SEQ ID NO: 12.
14. A nucleic acid encoding the antibody according to claim 1.
15. An expression cassette or vector comprising the nucleic acid according to claim 14 and a promoter operatively connected with said nucleic acid.
16. A host cell comprising the nucleic acid according to claim 14.
17. A conjugate comprising the antibody according to claim 1 conjugated to a further agent, wherein the further agent is a polypeptide or protein.
18. The conjugate according to claim 17, wherein the further agent is a cytokine, an immunomodulatory compound, a tumor-specific antibody or an immune checkpoint blocking or activating antibody.
19. A composition comprising the antibody according to claim 1.
20. (canceled)
21. A method for the treatment of cancer, an infection, an autoimmune disease or an immunodeficiency disorder, comprising the step of administering the antibody according to claim 1.
22. A method for the diagnosis, detecting and/or monitoring of cancer, an infection, an autoimmune disease or an immunodeficiency disorder, comprising the step of contacting a test sample with the antibody according to claim 1.
23. The method of treatment according to claim 21, wherein the cancer is characterized by expressing TA-MUC1.
24. The method of treatment according to claim 21, wherein the cancer is selected from the group consisting of ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemias, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
25. The method of treatment according to claim 21, wherein the antibody is used in combination with a further agent.
26. A method of increasing the MUC1 binding affinity of an antibody comprising
(i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
(ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6,
the method comprising the step of substituting the amino acid residue at position 8 of CDR-H2 with any amino acid residue except asparagine, resulting in CDR-H2 having the amino acid sequence of SEQ ID NO: 2.
27. The method according to claim 26, wherein substituting the amino acid residue at position 8 of CDR-H2 is achieved by introducing a mutation into the nucleic acid coding for the antibody, wherein the mutation is introduced in the codon coding for said amino acid residue.
28. A method of producing an antibody with increased MUC1 binding affinity, comprising
(a) providing a nucleic acid coding for an antibody which comprises
(i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 8 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
(ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6;
(b) introducing a mutation into said nucleic acid to produce a mutated nucleic acid, wherein the mutation is introduced in the codon coding for the amino acid residue at position 8 of CDR-H2 so that said codon codes for any amino acid residue except asparagine; and
(c) producing the antibody with increased MUC1 binding affinity by expressing the mutated nucleic acid in a host cell.
29. The method according to claim 28, wherein the antibody with increased MUC1 binding affinity comprises
(i) a heavy chain variable region comprising the complementarity-determining regions (CDRs) CDR-H1 having the amino acid sequence of SEQ ID NO: 1, CDR-H2 having the amino acid sequence of SEQ ID NO: 2 and CDR-H3 having the amino acid sequence of SEQ ID NO: 3, and
(ii) a light chain variable region comprising the complementarity-determining regions (CDRs) CDR-L1 having the amino acid sequence of SEQ ID NO: 4, CDR-L2 having the amino acid sequence of SEQ ID NO: 5 and CDR-L3 having the amino acid sequence of SEQ ID NO: 6.
30. The method according to claim 22, wherein the cancer is characterized by expressing TA-MUC1.
31. The method according to claim 22, wherein the cancer is selected from the group consisting of ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemias, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
32. The method according to claim 22, wherein the antibody is used in combination with a further agent.
33. A method for the treatment of cancer, an infection, an autoimmune disease or an immunodeficiency disorder, comprising the step of administering the conjugate according to claim 17.
34. The method of treatment according to claim 33, wherein the cancer is characterized by expressing TA-MUC1.
35. The method of treatment according to claim 33, wherein the cancer is selected from the group consisting of ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemias, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
36. The method of treatment according to claim 33, wherein the conjugate is used in combination with a further agent.
37. A method for the diagnosis, detecting and/or monitoring of cancer, an infection, an autoimmune disease or an immunodeficiency disorder, comprising the step of contacting a test sample with the conjugate according to claim 17.
38. The method according to claim 37, wherein the cancer is characterized by expressing TA-MUC1.
39. The method according to claim 37, wherein the cancer is selected from the group consisting of ovarian cancer, breast cancer, pancreatic cancer, lung cancer, colon cancer, stomach cancer, liver cancer, kidney cancer, blood cancer, endometrial cancer, thyroid cancer, leukemias, seminomas, melanomas, carcinomas, teratomas, lymphomas, sarcomas, mesotheliomas, neuroblastomas, gliomas, rectal cancer, adrenal cancer, skin cancer, cancer of the brain, cervical cancer, intestinal cancer, intestine cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, esophagus cancer, colorectal cancer, ear, nose and throat (ENT) cancer, prostate cancer, bladder cancer, cancer of the uterus and the metastases thereof.
40. The method according to claim 37, wherein the conjugate is used in combination with a further agent.
US17/055,303 2018-05-18 2019-05-17 Anti-muc1 antibody Pending US20210221910A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP18173253.8 2018-05-18
EP18173253 2018-05-18
PCT/EP2019/062756 WO2019219889A1 (en) 2018-05-18 2019-05-17 Anti-muc1 antibody

Publications (1)

Publication Number Publication Date
US20210221910A1 true US20210221910A1 (en) 2021-07-22

Family

ID=62217845

Family Applications (4)

Application Number Title Priority Date Filing Date
US17/055,323 Pending US20210187118A1 (en) 2018-05-18 2019-05-17 Anti-muc1 antibody-drug conjugate
US17/055,303 Pending US20210221910A1 (en) 2018-05-18 2019-05-17 Anti-muc1 antibody
US17/878,611 Active US11872289B2 (en) 2018-05-18 2022-08-01 Anti-MUC1 antibody-drug conjugate
US18/349,438 Pending US20230372520A1 (en) 2018-05-18 2023-07-10 Anti-muc1 antibody-drug conjugate

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US17/055,323 Pending US20210187118A1 (en) 2018-05-18 2019-05-17 Anti-muc1 antibody-drug conjugate

Family Applications After (2)

Application Number Title Priority Date Filing Date
US17/878,611 Active US11872289B2 (en) 2018-05-18 2022-08-01 Anti-MUC1 antibody-drug conjugate
US18/349,438 Pending US20230372520A1 (en) 2018-05-18 2023-07-10 Anti-muc1 antibody-drug conjugate

Country Status (21)

Country Link
US (4) US20210187118A1 (en)
EP (4) EP4249002A3 (en)
JP (3) JP7257422B2 (en)
KR (2) KR20210010565A (en)
CN (3) CN112449641A (en)
AU (2) AU2019270457A1 (en)
BR (2) BR112020023346A2 (en)
CA (2) CA3100745A1 (en)
CO (2) CO2020013664A2 (en)
DK (1) DK3794042T3 (en)
ES (1) ES2951674T3 (en)
HR (1) HRP20230787T1 (en)
HU (1) HUE063066T2 (en)
MX (2) MX2020011996A (en)
PH (2) PH12020551970A1 (en)
PL (1) PL3794041T3 (en)
RS (1) RS64379B1 (en)
SG (2) SG11202010493XA (en)
TW (2) TW202402806A (en)
WO (2) WO2019219891A1 (en)
ZA (1) ZA202006684B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023201234A3 (en) * 2022-04-12 2023-11-30 Minerva Biotechnologies Corporation Anti-variable muc1* antibodies and uses thereof
US11872289B2 (en) 2018-05-18 2024-01-16 Daiichi Sankyo Co., Ltd. Anti-MUC1 antibody-drug conjugate

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3108044A1 (en) * 2018-07-31 2020-02-06 Daiichi Sankyo Company, Limited Treatment of metastatic brain tumor by administration of antibody-drug conjugate
TW202116778A (en) 2019-07-10 2021-05-01 美商斯布雷克薩二號公司 Peptide conjugates of cytotoxins as therapeutics
PE20220485A1 (en) 2019-07-10 2022-04-04 Cybrexa 3 Inc PEPTIDE CONJUGATES OF MICROTUBULE-TARGETING AGENTS AS THERAPEUTICS
IL299153A (en) * 2020-07-13 2023-02-01 Regeneron Pharma Camptothecin analogs conjugated to a glutamine residue in a protein, and their use
CN113941007A (en) * 2020-07-16 2022-01-18 成都科岭源医药技术有限公司 Serial-connection double-medicine link assembly unit and application thereof
US11806405B1 (en) 2021-07-19 2023-11-07 Zeno Management, Inc. Immunoconjugates and methods
CN116059339B (en) * 2021-11-04 2023-09-22 元本(珠海横琴)生物科技有限公司 Medicine for treating and preventing cancer and application thereof
WO2023137026A1 (en) * 2022-01-12 2023-07-20 Regeneron Pharmaceuticals, Inc. Camptothecin analogs conjugated to a glutamine residue in a protein, and their use
WO2023209591A1 (en) 2022-04-27 2023-11-02 Daiichi Sankyo Company, Limited Combination of antibody-drug conjugate with ezh1 and/or ezh2 inhibitor
WO2024050524A1 (en) 2022-09-01 2024-03-07 University Of Georgia Research Foundation, Inc. Compositions and methods for directing apolipoprotein l1 to induce mammalian cell death

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090110632A1 (en) * 2005-09-23 2009-04-30 Robert Young Biological materials and uses thereof

Family Cites Families (191)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4966843A (en) 1982-11-01 1990-10-30 Cetus Corporation Expression of interferon genes in Chinese hamster ovary cells
AU2353384A (en) 1983-01-19 1984-07-26 Genentech Inc. Amplification in eukaryotic host cells
KR850004274A (en) 1983-12-13 1985-07-11 원본미기재 Method for preparing erythropoietin
US4943533A (en) 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
US4931275A (en) 1985-12-02 1990-06-05 Yeda Research & Development Co., Ltd. Anti-tumor vaccines and their preparation
US5683674A (en) 1987-01-07 1997-11-04 Imperial Cancer Research Technology Ltd. Antibody against human mucin core protein and method of preparing and using same
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
US5183884A (en) 1989-12-01 1993-02-02 United States Of America Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
US5637770A (en) 1991-01-16 1997-06-10 Daiichi Pharmaceutical Co., Ltd. Hexa-cyclic compound
JP3008226B2 (en) 1991-01-16 2000-02-14 第一製薬株式会社 Hexacyclic compounds
WO1992015682A1 (en) 1991-02-27 1992-09-17 Creative Biomolecules, Inc. Serine-rich peptide linkers
LU91067I2 (en) 1991-06-14 2004-04-02 Genentech Inc Trastuzumab and its variants and immunochemical derivatives including immotoxins
GB9200415D0 (en) 1992-01-09 1992-02-26 Bagshawe Kenneth D Inactivation of cytotoxic drugs
EP0636029B1 (en) 1992-04-13 2000-11-08 Dana-Farber Cancer Institute, Inc. Antibodies specific for carcinoma-associated antigens
JP3359955B2 (en) 1992-07-16 2002-12-24 第一製薬株式会社 Antitumor agent
US5804187A (en) 1992-11-16 1998-09-08 Cancer Research Fund Of Contra Costa Modified antibodies with human milk fat globule specificity
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
ATE301201T1 (en) 1993-06-07 2005-08-15 Vical Inc PLASMIDS USABLE FOR GENE THERAPY
DE4329004A1 (en) 1993-08-28 1995-03-09 Max Delbrueck Centrum Monoclonal antibodies against the Thomsen-Friedenreich antigen, their preparation and their use for tumour detection
AU7965694A (en) 1993-10-15 1995-05-04 Curators Of The University Of Missouri, The Dna encoding the human p2u receptor and null cells expressing p2u receptors
US5443953A (en) 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
US5997873A (en) 1994-01-13 1999-12-07 Mount Sinai School Of Medicine Of The City University Of New York Method of preparation of heat shock protein 70-peptide complexes
US5961979A (en) 1994-03-16 1999-10-05 Mount Sinai School Of Medicine Of The City University Of New York Stress protein-peptide complexes as prophylactic and therapeutic vaccines against intracellular pathogens
US5888773A (en) 1994-08-17 1999-03-30 The United States Of America As Represented By The Department Of Health And Human Services Method of producing single-chain Fv molecules
JPH08337584A (en) 1995-04-10 1996-12-24 Dai Ichi Seiyaku Co Ltd Condensed six-membered cyclic amino compound, medicine containing the same and production of the same
US6504029B1 (en) 1995-04-10 2003-01-07 Daiichi Pharmaceutical Co., Ltd. Condensed-hexacyclic compounds and a process therefor
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
EP0831880A4 (en) 1995-06-07 2004-12-01 Imclone Systems Inc Antibody and antibody fragments for inhibiting the growth of tumors
CA2225460A1 (en) 1995-06-23 1997-01-09 Winston Campbell Patterson Transcriptional regulation of genes encoding vascular endothelial growth factor receptors
SG50747A1 (en) 1995-08-02 1998-07-20 Tanabe Seiyaku Co Comptothecin derivatives
JPH1095802A (en) 1995-12-28 1998-04-14 Tanabe Seiyaku Co Ltd Camptothecine derivative
CA2192725C (en) 1995-12-28 2004-04-20 Kenji Tsujihara Camptothecin derivatives
GB9603256D0 (en) 1996-02-16 1996-04-17 Wellcome Found Antibodies
US5968511A (en) 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
EP0906444A1 (en) 1996-04-19 1999-04-07 Gabriele Pecher Genetically transfected human dendritic cells, their production and use, preferably as vaccines
TW409058B (en) 1996-06-06 2000-10-21 Daiichi Seiyaku Co Method for preparation of a drug complex
TW527183B (en) 1996-06-06 2003-04-11 Daiichi Seiyaku Co Drug complex
JPH1171280A (en) 1997-06-26 1999-03-16 Tanabe Seiyaku Co Ltd Medicine composition
US6172213B1 (en) 1997-07-02 2001-01-09 Genentech, Inc. Anti-IgE antibodies and method of improving polypeptides
JPH1192405A (en) 1997-09-19 1999-04-06 Dai Ichi Seiyaku Co Ltd Medicinal complex
EP1987845B1 (en) 1997-11-20 2012-03-21 Vical Incorporated Treatment of cancer using cytokine-expressing polynucleotides and compositions therefor.
US5948646A (en) 1997-12-11 1999-09-07 Fordham University Methods for preparation of vaccines against cancer comprising heat shock protein-peptide complexes
DK1071700T3 (en) 1998-04-20 2010-06-07 Glycart Biotechnology Ag Glycosylation modification of antibodies to enhance antibody-dependent cellular cytotoxicity
CA2333321A1 (en) 1998-05-22 1999-12-02 Daiichi Pharmaceutical Co., Ltd. Drug complex
ID29943A (en) 1998-10-30 2001-10-25 Daiichi Seiyaku Co COMPOUND DDS AND MEASUREMENT METHODS
US6465220B1 (en) 1998-12-21 2002-10-15 Glycozym Aps Glycosylation using GalNac-T4 transferase
AUPP816899A0 (en) 1999-01-14 1999-02-11 Food Technology Innovations Pty Limited Improved microbial products
WO2000052135A2 (en) 1999-03-02 2000-09-08 Human Genome Sciences, Inc. Engineering intracellular sialylation pathways
WO2000058499A1 (en) 1999-03-30 2000-10-05 Japan Tobacco Inc. Process for producing monoclonal antibody
DK2270150T4 (en) 1999-04-09 2019-08-26 Kyowa Hakko Kirin Co Ltd PROCEDURE TO CONTROL THE ACTIVITY OF IMMUNOLOGICAL FUNCTIONAL MOLECULE.
JP4780633B2 (en) 1999-06-25 2011-09-28 イムノゲン インコーポレーティッド Method of treatment using anti-ErbB antibody-maytansinoid complex
WO2001012217A1 (en) 1999-08-18 2001-02-22 Altarex Corp. Therapeutic antibody against muc-1 antigen and methods for their use
US7147850B2 (en) 1999-08-18 2006-12-12 Altarex Medical Corp. Therapeutic binding agents against MUC-1 antigen and methods for their use
AU7743100A (en) 1999-09-30 2001-04-30 Corixa Corporation Stress protein compositions and methods for prevention and treatment of cancer and infectious disease
JP2002060351A (en) 2000-03-22 2002-02-26 Dai Ichi Seiyaku Co Ltd Dds compound comprising drug having hydroxy group
CA2403998A1 (en) 2000-03-30 2001-10-11 Dyax Corp. Mucin-1 specific binding members and methods of use thereof
GB2362531A (en) 2000-05-15 2001-11-21 Nokia Mobile Phones Ltd Indicating the temporal order of reference frames in a video sequence
FR2809312B1 (en) 2000-05-25 2002-07-12 Gervais Danone Sa USE OF L. CASEI IN IMMUNOSTIMULATING COMPOSITIONS
IL153505A0 (en) 2000-06-29 2003-07-06 Daiichi Seiyaku Co Dds compound and process for the preparation thereof
CN1556712A (en) 2000-07-13 2004-12-22 ��һ��������ҩ��ʽ���� Pharmaceutical compositions containing DDS compounds
FR2814096B1 (en) 2000-09-15 2002-12-27 Montupet Sa METHOD OF MANUFACTURING FOUNDRY PIECES EQUIPPED WITH INSERTS WITH IMPROVED PIECE / INSERT MECHANICAL COHESION, AND INSERT THAT CAN BE USED IN SUCH A PROCESS
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
JPWO2002030954A1 (en) 2000-10-06 2004-02-19 協和醗酵工業株式会社 Methods for purifying antibodies
GB0029360D0 (en) 2000-12-01 2001-01-17 Univ Nottingham Humanised antibodies and uses thereof
KR100866199B1 (en) 2001-02-01 2008-10-30 가부시끼가이샤 야구르트혼샤 Method of evaluating the extent of reaching the intestine of bifidobacterium in fermented milk food or drink
US7183388B2 (en) 2001-03-30 2007-02-27 The Regents Of The University Of California Anti-MUC-1 single chain antibodies for tumor targeting
EP1283053A1 (en) 2001-08-09 2003-02-12 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Inhibitors of HER3 activity
US20050042714A1 (en) 2001-08-17 2005-02-24 Michael Pawlita Glycoconjugates of sialic acid derivates, methods for their production and use thereof
CA2451998A1 (en) 2001-08-17 2003-02-27 Eli Lilly And Company Anti-a.beta. antibodies
DE10139428A1 (en) 2001-08-17 2003-03-27 Nemod Immuntherapie Ag Preparation of dendritic cells, useful e.g. as antitumor or antimicrobial vaccines, by treating CD124- and CD116-positive cells with stimulatory molecules
TWI313609B (en) 2001-08-21 2009-08-21 Mitsubishi Tanabe Pharma Corp Pharmaceutical composition for inhibiting the metastasis or preventing the recurrence of malignant tumor
DE60212425T2 (en) 2001-10-22 2006-11-09 Applied Research Systems Ars Holding N.V. FSH COMPOSITIONS HAVING A HIGH SIALYLATION RATE AND THEIR USE FOR THE PREPARATION OF MEDICAMENTS
GB0125473D0 (en) 2001-10-24 2001-12-12 Syngenta Ltd Chemical process
KR100988949B1 (en) 2001-10-25 2010-10-20 제넨테크, 인크. Glycoprotein compositions
CA2468088A1 (en) 2001-11-20 2003-05-30 Atgen Co., Ltd. Novel peptides conferring environmental stress resistance and fusion proteins including said peptides
CA2467242A1 (en) 2001-11-20 2003-05-30 Seattle Genetics, Inc. Treatment of immunological disorders using anti-cd30 antibodies
US8877901B2 (en) 2002-12-13 2014-11-04 Immunomedics, Inc. Camptothecin-binding moiety conjugates
AU2003217912A1 (en) 2002-03-01 2003-09-16 Xencor Antibody optimization
HUE054986T2 (en) 2002-03-01 2021-10-28 Immunomedics Inc Immunoconjugate comprising humanised rs7 antibodies
US9745380B2 (en) 2002-03-01 2017-08-29 Immunomedics, Inc. RS7 antibodies
GB0212046D0 (en) 2002-05-24 2002-07-03 Glaxo Group Ltd Vaccines
AU2003266236A1 (en) 2002-07-22 2004-02-09 Nemod Immuntherapie Ag Method for the production of an immunostimulating mucin (muc1)
ATE482720T1 (en) 2002-08-16 2010-10-15 Glycotope Gmbh METHOD FOR PRODUCING TEMPERATURE-INDUCED TUMOR CELL LYSATES FOR USE AS IMMUNOGENIC COMPOUNDS
DE10256900A1 (en) 2002-11-29 2004-06-24 Nemod Immuntherapie Ag Tumor-specific recognition molecules
WO2004054622A1 (en) 2002-12-13 2004-07-01 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
DE10303664A1 (en) * 2003-01-23 2004-08-12 Nemod Immuntherapie Ag Detection molecules for the treatment and detection of tumors
US20040202666A1 (en) 2003-01-24 2004-10-14 Immunomedics, Inc. Anti-cancer anthracycline drug-antibody conjugates
US7709610B2 (en) 2003-05-08 2010-05-04 Facet Biotech Corporation Therapeutic use of anti-CS1 antibodies
WO2005019258A2 (en) 2003-08-11 2005-03-03 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
ES2412489T3 (en) 2003-08-14 2013-07-11 Thrombogenics N.V. Antibodies against factor VIII with modified glycosylation in the variable region
ES2425475T3 (en) 2003-08-18 2013-10-15 Glycotope Gmbh NM-F9 (DSM ACC2606) and NM-D4 (DSM ACC2605) tumor cell lines, uses thereof
FR2861080B1 (en) 2003-10-20 2006-02-17 Lab Francais Du Fractionnement ANTIBODIES HAVING AN OPTIMIZED FUCOSE AND GALACTOSE RATE
CA2557725C (en) 2004-02-13 2015-06-30 Glycotope Gmbh Highly active glycoproteins-process conditions and an efficient method for their production
CN1964716A (en) 2004-02-26 2007-05-16 伊诺泰克制药公司 Isoquinoline derivatives and methods of use thereof
EP1725586B1 (en) 2004-03-02 2015-01-14 Seattle Genetics, Inc. Partially loaded antibodies and methods of their conjugation
ES2393539T3 (en) 2004-03-29 2012-12-26 Abbott Biotherapeutics Corp. Therapeutic use of anti-CS1 antibodies
WO2005108423A1 (en) 2004-05-11 2005-11-17 Atgen Co., Ltd. Novel peptides conferring environmental stress resistance and fusion proteins including said peptides
US20060051353A1 (en) 2004-05-14 2006-03-09 Jean-Frederic Colombel Methods and compositions to evaluate antibody treatment response
AU2005244980B2 (en) 2004-05-19 2011-09-15 E. R. Squibb & Sons, L.L.C. Chemical linkers and conjugates thereof
MXPA06014065A (en) 2004-06-01 2007-01-31 Genentech Inc Antibody drug conjugates and methods.
ZA200701234B (en) 2004-07-22 2008-12-31 Genentech Inc HER2 antibody composition
US7374755B2 (en) 2004-07-26 2008-05-20 The Research Foundation Of State University Of New York Therapeutic use of anti-TF-Antigen antibody
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
EP1817341A2 (en) 2004-11-29 2007-08-15 Seattle Genetics, Inc. Engineered antibodies and immunoconjugates
CA2592177A1 (en) 2005-01-21 2006-07-27 Genentech, Inc. Fixed dosing of her antibodies
EP1848804B1 (en) * 2005-01-28 2011-10-05 Ramot at Tel Aviv University, Ltd. Anti-muc1 alpha beta antibodies
DE102005009099A1 (en) 2005-02-28 2006-08-31 Ktb Tumorforschungsgesellschaft Mbh New camptothecin-peptide derivatives, useful for treating cancer
DE102005009084A1 (en) 2005-02-28 2006-08-31 Ktb Tumorforschungsgesellschaft Mbh New anthracyclin-peptide derivatives, useful for treating cancer, especially of the prostate, are cleaved, in the tumor, by prostate-specific antigen to release active antitumor agent and are transported by serum albumen
US7833979B2 (en) 2005-04-22 2010-11-16 Amgen Inc. Toxin peptide therapeutic agents
WO2006125207A2 (en) 2005-05-19 2006-11-23 Amgen Inc. Compositions and methods for increasing the stability of antibodies
ES2543685T3 (en) 2005-06-30 2015-08-21 Janssen Biotech, Inc. Methods and compositions with improved therapeutic activity
JP4919146B2 (en) * 2005-09-27 2012-04-18 独立行政法人産業技術総合研究所 Switching element
CA2631630A1 (en) 2005-11-30 2007-06-07 University Of Southern California Fc.gamma. polymorphisms for predicting disease and treatment outcome
AR056857A1 (en) 2005-12-30 2007-10-24 U3 Pharma Ag DIRECTED ANTIBODIES TO HER-3 (RECEIVER OF THE HUMAN EPIDERMAL GROWTH FACTOR-3) AND ITS USES
JP2009527066A (en) 2006-02-14 2009-07-23 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ Bit detection for optical disk reading
US20080131428A1 (en) 2006-02-24 2008-06-05 Arius Research, Inc. Cytotoxicity mediation of cells evidencing surface expression of TROP-2
AR060070A1 (en) 2006-03-24 2008-05-21 Merck Patent Gmbh HETERODYMERIC PROTEIN DOMAINS OBTAINED BY ENGINEERING
WO2007124992A1 (en) 2006-04-28 2007-11-08 Unilever N.V. Method of manufacturing a cultured edible product comprising omega-3 polyunsaturated fatty acids
US8524865B2 (en) 2006-05-30 2013-09-03 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
SI2032606T1 (en) 2006-05-30 2014-02-28 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
ITMI20061473A1 (en) 2006-07-26 2008-01-27 Indena Spa DERIVATIVES OF CAMPTOTECIN WITH ANTITUMORAL ACTIVITY
EP1911766A1 (en) 2006-10-13 2008-04-16 Glycotope Gmbh Use of human cells of myeloid leukaemia origin for expression of antibodies
EP1900750A1 (en) 2006-09-18 2008-03-19 Glycotope Gmbh Fully human high yield production system for improved antibodies
EP3539981A1 (en) 2006-09-10 2019-09-18 Glycotope GmbH Use of human cells of myeloid leukaemia origin for expression of antibodies
EP1920781B1 (en) 2006-11-10 2015-03-04 Glycotope GmbH Compositions comprising a core-1 positive microorganism and their use for the treatment or prophylaxis of tumors
CN101687838A (en) 2006-12-21 2010-03-31 先灵公司 Suppress the active pyrrolo-of KSP kinesin [3,2-A] pyridine derivate
PT2129396E (en) 2007-02-16 2013-11-18 Merrimack Pharmaceuticals Inc Antibodies against erbb3 and uses thereof
WO2008101177A2 (en) 2007-02-16 2008-08-21 University Of Virginia Patent Foundation Ige antibodies to chimeric or humanized igg therapeutic monoclonal antibodies as a screening test for anaphylaxis
CA2681159A1 (en) 2007-03-13 2008-09-18 Peros Systems Technologies, Inc. Immunoactive compositions for improved oral delivery of vaccines and therapeutic agents
KR20100014527A (en) 2007-03-22 2010-02-10 슬로안-케테링인스티튜트퍼캔서리서치 Uses of monoclonal antibody 8h9
CN101687915B8 (en) 2007-04-03 2018-08-03 安进研发(慕尼黑)股份有限公司 Cross-species-specific cd 3-epsilon binding domain
DK2281006T3 (en) 2008-04-30 2017-11-06 Immunogen Inc CROSS-BONDING AGENTS AND APPLICATIONS THEREOF
WO2009140242A1 (en) 2008-05-13 2009-11-19 Genentech, Inc. Analysis of antibody drug conjugates by bead-based affinity capture and mass spectrometry
AU2009248410B2 (en) 2008-05-13 2014-07-24 Glycotope Gmbh Fermentation process
CN102112494A (en) 2008-06-03 2011-06-29 雅培制药有限公司 Dual variable domain immunoglobulins and uses thereof
CN102264765B (en) 2008-10-28 2016-01-20 盐野义制药株式会社 Anti-MUC1 antibody
CN102282168A (en) 2008-11-18 2011-12-14 梅里麦克制药股份有限公司 Human serum albumin linkers and conjugates thereof
PL2396036T3 (en) 2009-02-13 2017-12-29 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US9079957B2 (en) 2009-04-16 2015-07-14 The University Of Tokyo Diagnosis and treatment of cancer using anti-TMPRSS11E antibody
JP2013500253A (en) 2009-07-22 2013-01-07 エンゾン ファーマシューティカルズ,インコーポレーテッド Method for treating HER2-positive cancer using HER2 receptor antagonist combined with multi-arm polymer complex of 7-ethyl-10-hydroxycamptothecin
EP2281844A1 (en) 2009-07-31 2011-02-09 Glycotope GmbH MUC 1 antibodies
WO2011021397A1 (en) 2009-08-20 2011-02-24 国立大学法人千葉大学 Colchicine derivatives
US20110070248A1 (en) 2009-09-24 2011-03-24 Seattle Genetics, Inc. Dr5 ligand drug conjugates
RS60390B1 (en) 2009-11-13 2020-07-31 Daiichi Sankyo Europe Gmbh Material and methods for treating or preventing her-3 associated diseases
WO2011068845A1 (en) 2009-12-02 2011-06-09 Immunomedics, Inc. Combining radioimmunotherapy and antibody-drug conjugates for improved cancer therapy
ES2701626T3 (en) 2009-12-28 2019-02-25 Oncotherapy Science Inc Anti-CDH3 antibodies and their uses
EP2347769A1 (en) 2010-01-20 2011-07-27 Glycotope GmbH Cancer stem cell markers and uses thereof
US9127088B2 (en) 2010-05-13 2015-09-08 Indiana University Research And Technology Corporation Glucagon superfamily peptides exhibiting nuclear hormone receptor activity
BR112012029281B1 (en) 2010-05-17 2020-12-08 Livtech, Inc antibody to human trop-2, monoclonal antibody to human trop-2, antibody-derived antibody fragment, hybridoma, conjugate, pharmaceutical composition, use of pharmaceutical composition, method to detect a tumor and kit to treat, diagnose or detect a tumor
JP5881254B2 (en) 2010-05-18 2016-03-09 セルリアン・ファーマ・インコーポレイテッド Compositions and methods for the treatment of autoimmune and other diseases
EP2594589A1 (en) 2010-06-10 2013-05-22 Sapporo Medical University ANTI-Trop-2 ANTIBODY
EP2593142B8 (en) 2010-07-12 2018-12-26 Pfizer Healthcare Ireland Multifunctional antibody conjugates
WO2012019024A2 (en) 2010-08-04 2012-02-09 Immunogen, Inc. Her3-binding molecules and immunoconjugates thereof
US20120156130A1 (en) 2010-08-06 2012-06-21 Thore Hettmann Use of her3 binding agents in prostate treatment
US9051370B2 (en) 2010-08-10 2015-06-09 Glycotope Gmbh Humanized EGFR antibodies
EP2603528B1 (en) 2010-08-10 2016-10-12 Glycotope GmbH Fab-glycosylated antibodies
WO2012064733A2 (en) 2010-11-09 2012-05-18 Medimmune, Llc Antibody scaffold for homogenous conjugation
US20120121615A1 (en) 2010-11-17 2012-05-17 Flygare John A Alaninyl maytansinol antibody conjugates
US9175092B2 (en) 2011-06-28 2015-11-03 Oxford Biotherapeutics Ltd Antibodies to bone marrow stromal antigen 1
BR112014011331A2 (en) 2011-11-11 2017-04-25 Rinat Neuroscience Corp trop-2 specific antibodies and their uses
US9427464B2 (en) 2011-11-22 2016-08-30 Chiome Bioscience Inc. Anti-human TROP-2 antibody having an antitumor activity in vivo
EP2790731A2 (en) 2011-12-14 2014-10-22 Seattle Genetics, Inc. Fgfr-binder-active agent conjugates
BR112014015111A2 (en) 2011-12-21 2017-06-13 Novartis Ag p-factor targeting antibody compositions and processes
US20130280282A1 (en) 2012-04-24 2013-10-24 Daiichi Sankyo Co., Ltd. Dr5 ligand drug conjugates
JP2015521602A (en) 2012-06-14 2015-07-30 アンブルックス, インコーポレイテッドAmbrx, Inc. Anti-PSMA antibodies conjugated to nuclear receptor ligand polypeptides
KR102369419B1 (en) 2012-10-11 2022-03-02 다이이찌 산쿄 가부시키가이샤 Method for producing a glycinamide compound
EP2910573B1 (en) 2012-10-19 2020-02-19 Daiichi Sankyo Company, Limited Antibody-drug conjugate produced by binding through linker having hydrophilic structure
KR102643443B1 (en) 2012-11-13 2024-03-06 비온테크 에스이 Agents for treatment of claudin expressing cancer diseases
EP2927227A4 (en) 2013-01-03 2015-12-30 Celltrion Inc Antibody-linker-drug conjugate, preparation method therefor, and anticancer drug composition containing same
EP2945964A2 (en) 2013-01-18 2015-11-25 Glycotope GmbH Fusion peptides for enhancing protein expression
EA201690321A1 (en) * 2013-08-02 2016-12-30 Санофи APPLICATION OF ANTIBODY IMMUNOCONG CONTRACT TO MUC1 AND MAITANZINOID FOR THE TREATMENT OF SOLID TUMORS
MX2016007972A (en) 2013-12-17 2016-10-28 Genentech Inc Methods of treating cancers using pd-1 axis binding antagonists and taxanes.
KR20230078820A (en) 2013-12-25 2023-06-02 다이이찌 산쿄 가부시키가이샤 Anti-trop2 antibody-drug conjugate
JP2015138634A (en) 2014-01-22 2015-07-30 株式会社オートネットワーク技術研究所 Electric wire with terminal fitting and manufacturing method thereof
JP6908381B2 (en) * 2014-01-29 2021-07-28 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Antibodies to MUC1-C / extracellular domain (MUC1-C / ECD)
BR112016013482B1 (en) 2014-01-31 2022-04-19 Daiichi Sankyo Company, Limited ANTI-HER2-DRUG ANTIBODY CONJUGATE, ANTI-TUMOR AND/OR ANTI-CANCER DRUGS AND PHARMACEUTICAL COMPOSITION
IL311108A (en) 2014-04-10 2024-04-01 Daiichi Sankyo Co Ltd Method for producing an anti-her3 antibody-drug conjugate
GB201406767D0 (en) * 2014-04-15 2014-05-28 Cancer Rec Tech Ltd Humanized anti-Tn-MUC1 antibodies anf their conjugates
CA2947404C (en) 2014-04-28 2023-05-16 Medicinal Chemistry Pharmaceuticals, Co., Ltd. Anti-muc1 antibody or antigen-binding fragment thereof and uses thereof
CN106659775B (en) 2014-06-30 2021-08-06 阿尔托生物科学有限公司 IL-15-based molecules, methods and uses thereof
LU92659B1 (en) 2015-02-23 2016-08-24 Glycotope Gmbh Glycooptimized antibody drug conjugates
SG11201707541WA (en) 2015-03-17 2017-10-30 Tilt Biotherapeutics Oy Oncolytic adenoviruses coding for bi-specific antibodies and methods and uses related thereto
US10524738B2 (en) 2015-05-04 2020-01-07 Cercacor Laboratories, Inc. Noninvasive sensor system with visual infographic display
EP3574015A1 (en) 2017-01-27 2019-12-04 Glycotope GmbH Anti-cancer treatments with an anti-muc1 antibody and an erbb inhibitor
CN111315776A (en) 2017-03-29 2020-06-19 葛莱高托普有限公司 PD-L1 and TA-MUC1 antibodies
KR20190134994A (en) 2017-03-29 2019-12-05 글리코토페 게엠베하 Multispecific Antibody Constructs That Bind to MUC1 and CD3
CN110382541A (en) 2017-03-29 2019-10-25 葛莱高托普有限公司 Humanization anti-CD 40 antibodies
WO2018178123A1 (en) 2017-03-29 2018-10-04 Glycotope Gmbh BISPECIFIC MUC-1 x PD-L1 ANTIBODIES
US10561686B2 (en) 2018-01-12 2020-02-18 Innovative Cellular Therapeutics CO., LTD. Modified cell expansion and uses thereof
EP3759144A1 (en) 2018-03-01 2021-01-06 Glycotope GmbH Fusion protein constructs comprising an anti-muc1 antibody and il-15
SG11202010493XA (en) 2018-05-18 2020-11-27 Glycotope Gmbh Anti-muc1 antibody

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090110632A1 (en) * 2005-09-23 2009-04-30 Robert Young Biological materials and uses thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11872289B2 (en) 2018-05-18 2024-01-16 Daiichi Sankyo Co., Ltd. Anti-MUC1 antibody-drug conjugate
WO2023201234A3 (en) * 2022-04-12 2023-11-30 Minerva Biotechnologies Corporation Anti-variable muc1* antibodies and uses thereof

Also Published As

Publication number Publication date
US20210187118A1 (en) 2021-06-24
PH12020551969A1 (en) 2021-09-20
JP2021524740A (en) 2021-09-16
AU2019270459A1 (en) 2020-12-03
EP3794041B1 (en) 2023-07-12
CA3100608A1 (en) 2019-11-21
CA3100745A1 (en) 2019-11-21
SG11202010493XA (en) 2020-11-27
JP2023027259A (en) 2023-03-01
EP3794042A1 (en) 2021-03-24
EP4249002A2 (en) 2023-09-27
AU2019270457A1 (en) 2020-12-03
ZA202006684B (en) 2021-09-29
WO2019219891A1 (en) 2019-11-21
PH12020551970A1 (en) 2021-09-13
CO2020014435A2 (en) 2020-12-21
JP2021524852A (en) 2021-09-16
EP4257611A2 (en) 2023-10-11
CN117815404A (en) 2024-04-05
CO2020013664A2 (en) 2020-12-21
MX2020011996A (en) 2021-01-29
US20230139769A1 (en) 2023-05-04
US11872289B2 (en) 2024-01-16
RS64379B1 (en) 2023-08-31
EP4249002A3 (en) 2023-11-22
TWI825098B (en) 2023-12-11
US20230372520A1 (en) 2023-11-23
HRP20230787T1 (en) 2023-10-27
DK3794042T3 (en) 2024-04-15
MX2020011997A (en) 2021-03-25
KR20210013107A (en) 2021-02-03
EP3794041A1 (en) 2021-03-24
KR20210010565A (en) 2021-01-27
TW202402806A (en) 2024-01-16
CN112351999A (en) 2021-02-09
ES2951674T3 (en) 2023-10-24
BR112020023373A2 (en) 2021-02-09
EP3794041C0 (en) 2023-07-12
SG11202010496WA (en) 2020-12-30
HUE063066T2 (en) 2024-01-28
EP4257611A3 (en) 2024-03-06
PL3794041T3 (en) 2023-08-28
TW202003583A (en) 2020-01-16
CN112449641A (en) 2021-03-05
JP7257422B2 (en) 2023-04-13
EP3794042B1 (en) 2024-03-27
WO2019219889A1 (en) 2019-11-21
TW202003579A (en) 2020-01-16
BR112020023346A2 (en) 2021-02-09

Similar Documents

Publication Publication Date Title
EP3794041B1 (en) Anti-muc1 antibody
US20160376368A1 (en) Antibodies to integrin avb6 and use of same to treat cancer
CN107880126B (en) anti-NTB-A antibodies and related compositions and methods
JP6828056B2 (en) Anti-HER2 antibody-drug conjugate and its use
US20220267467A1 (en) Polypeptide complex for conjugation and use thereof
RU2792347C2 (en) Antibody against muc1
CA3055433A1 (en) Humanized anti-cd40 antibodies
TW202221034A (en) Anti-cd228 antibodies and antibody-drug conjugates
RU2804703C2 (en) Anti-muc1 drug conjugate
US20230235076A1 (en) Humanized antibodies against lewis y
WO2023175117A1 (en) Antibodies against lypd3

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLYCOTOPE GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GELLERT, JOHANNA;FLECHNER, ANKE;WEIGELT, DOREEN;AND OTHERS;REEL/FRAME:054883/0196

Effective date: 20201110

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED